134 results on '"Mahmoud A. Pouladi"'
Search Results
2. CHCHD2 up-regulation in Huntington disease mediates a compensatory protective response against oxidative stress
- Author
-
Xuanzhuo Liu, Fang Wang, Xinman Fan, Mingyi Chen, Xiaoxin Xu, Qiuhong Xu, Huili Zhu, Anding Xu, Mahmoud A. Pouladi, and Xiaohong Xu
- Subjects
Cytology ,QH573-671 - Abstract
Abstract Huntington disease (HD) is a neurodegenerative disease caused by the abnormal expansion of a polyglutamine tract resulting from a mutation in the HTT gene. Oxidative stress has been identified as a significant contributing factor to the development of HD and other neurodegenerative diseases, and targeting anti-oxidative stress has emerged as a potential therapeutic approach. CHCHD2 is a mitochondria-related protein involved in regulating cell migration, anti-oxidative stress, and anti-apoptosis. Although CHCHD2 is highly expressed in HD cells, its specific role in the pathogenesis of HD remains uncertain. We postulate that the up-regulation of CHCHD2 in HD models represents a compensatory protective response against mitochondrial dysfunction and oxidative stress associated with HD. To investigate this hypothesis, we employed HD mouse striatal cells and human induced pluripotent stem cells (hiPSCs) as models to examine the effects of CHCHD2 overexpression (CHCHD2-OE) or knockdown (CHCHD2-KD) on the HD phenotype. Our findings demonstrate that CHCHD2 is crucial for maintaining cell survival in both HD mouse striatal cells and hiPSCs-derived neurons. Our study demonstrates that CHCHD2 up-regulation in HD serves as a compensatory protective response against oxidative stress, suggesting a potential anti-oxidative strategy for the treatment of HD.
- Published
- 2024
- Full Text
- View/download PDF
3. Claudin-11 in health and disease: implications for myelin disorders, hearing, and fertility
- Author
-
Sophia C. Gjervan, Oguz K. Ozgoren, Alexander Gow, Sylvia Stockler-Ipsiroglu, and Mahmoud A. Pouladi
- Subjects
claudin-11 ,tight junctions ,myelin ,leukodystrophy ,HLD22 ,hearing ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Claudin-11 plays a critical role in multiple physiological processes, including myelination, auditory function, and spermatogenesis. Recently, stop-loss mutations in CLDN11 have been identified as a novel cause of hypomyelinating leukodystrophy (HLD22). Understanding the multifaceted roles of claudin-11 and the potential pathogenic mechanisms in HLD22 is crucial for devising targeted therapeutic strategies. This review outlines the biological roles of claudin-11 and the implications of claudin-11 loss in the context of the Cldn11 null mouse model. Additionally, HLD22 and proposed pathogenic mechanisms, such as endoplasmic reticulum stress, will be discussed.
- Published
- 2024
- Full Text
- View/download PDF
4. Generation of a human induced pluripotent stem cell line from a patient with hypomyelinating leukodystrophy 22 (HLD22)
- Author
-
Oguz K. Ozgoren, Glen Lester Sequiera, Costanza Ferrari Bardile, Sophia C. Gjervan, Areesha Salman, Anna Lehman, Stuart E. Turvey, Colin J.D. Ross, Sylvia Stockler, and Mahmoud A. Pouladi
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Hypomyelinating Leukodystrophy 22 (HLD22) is caused by a stoploss mutation in CLDN11. To study the molecular mechanisms underlying HLD22, human induced pluripotent stem cells (hiPSCs) were generated from patient fibroblasts carrying the stop-loss mutation in CLDN11.
- Published
- 2023
- Full Text
- View/download PDF
5. Widespread dysregulation of mRNA splicing implicates RNA processing in the development and progression of Huntington's diseaseResearch in context
- Author
-
Vincent Tano, Kagistia Hana Utami, Nur Amirah Binte Mohammad Yusof, Jocelyn Bégin, Willy Wei Li Tan, Mahmoud A. Pouladi, and Sarah R. Langley
- Subjects
Huntington's disease ,Alternative splicing ,mRNA processing ,Neurodegenerative disease ,High-throughput RNA-sequencing ,Proteomics ,Medicine ,Medicine (General) ,R5-920 - Abstract
Summary: Background: In Huntington's disease (HD), a CAG repeat expansion mutation in the Huntingtin (HTT) gene drives a gain-of-function toxicity that disrupts mRNA processing. Although dysregulation of gene splicing has been shown in human HD post-mortem brain tissue, post-mortem analyses are likely confounded by cell type composition changes in late-stage HD, limiting the ability to identify dysregulation related to early pathogenesis. Methods: To investigate gene splicing changes in early HD, we performed alternative splicing analyses coupled with a proteogenomics approach to identify early CAG length-associated splicing changes in an established isogenic HD cell model. Findings: We report widespread neuronal differentiation stage- and CAG length-dependent splicing changes, and find an enrichment of RNA processing, neuronal function, and epigenetic modification-related genes with mutant HTT-associated splicing. When integrated with a proteomics dataset, we identified several of these differential splicing events at the protein level. By comparing with human post-mortem and mouse model data, we identified common patterns of altered splicing from embryonic stem cells through to post-mortem striatal tissue. Interpretation: We show that widespread splicing dysregulation in HD occurs in an early cell model of neuronal development. Importantly, we observe HD-associated splicing changes in our HD cell model that were also identified in human HD striatum and mouse model HD striatum, suggesting that splicing-associated pathogenesis possibly occurs early in neuronal development and persists to later stages of disease. Together, our results highlight splicing dysregulation in HD which may lead to disrupted neuronal function and neuropathology. Funding: This research is supported by the Lee Kong Chian School of Medicine, Nanyang Technological University Singapore Nanyang Assistant Professorship Start-Up Grant, the Singapore Ministry of Education under its Singapore Ministry of Education Academic Research Fund Tier 1 (RG23/22), the BC Children's Hospital Research Institute Investigator Grant Award (IGAP), and a Scholar Award from the Michael Smith Health Research BC.
- Published
- 2023
- Full Text
- View/download PDF
6. Pluripotent stem cell-derived models of neurological diseases reveal early transcriptional heterogeneity
- Author
-
Matan Sorek, Walaa Oweis, Malka Nissim-Rafinia, Moria Maman, Shahar Simon, Cynthia C. Hession, Xian Adiconis, Sean K. Simmons, Neville E. Sanjana, Xi Shi, Congyi Lu, Jen Q. Pan, Xiaohong Xu, Mahmoud A. Pouladi, Lisa M. Ellerby, Feng Zhang, Joshua Z. Levin, and Eran Meshorer
- Subjects
Transcriptional heterogeneity ,Single cell ,scRNA-seq ,Neurological diseases ,Neurodegenerative diseases ,Huntington’s disease ,Biology (General) ,QH301-705.5 ,Genetics ,QH426-470 - Abstract
Abstract Background Many neurodegenerative diseases develop only later in life, when cells in the nervous system lose their structure or function. In many forms of neurodegenerative diseases, this late-onset phenomenon remains largely unexplained. Results Analyzing single-cell RNA sequencing from Alzheimer’s disease (AD) and Huntington’s disease (HD) patients, we find increased transcriptional heterogeneity in disease-state neurons. We hypothesize that transcriptional heterogeneity precedes neurodegenerative disease pathologies. To test this idea experimentally, we use juvenile forms (72Q; 180Q) of HD iPSCs, differentiate them into committed neuronal progenitors, and obtain single-cell expression profiles. We show a global increase in gene expression variability in HD. Autophagy genes become more stable, while energy and actin-related genes become more variable in the mutant cells. Knocking down several differentially variable genes results in increased aggregate formation, a pathology associated with HD. We further validate the increased transcriptional heterogeneity in CHD8+/− cells, a model for autism spectrum disorder. Conclusions Overall, our results suggest that although neurodegenerative diseases develop over time, transcriptional regulation imbalance is present already at very early developmental stages. Therefore, an intervention aimed at this early phenotype may be of high diagnostic value.
- Published
- 2021
- Full Text
- View/download PDF
7. Elevated de novo protein synthesis in FMRP-deficient human neurons and its correction by metformin treatment
- Author
-
Kagistia Hana Utami, Nur Amirah Binte Mohammad Yusof, Jing Eugene Kwa, Ulla-Kaisa Peteri, Maija L. Castrén, and Mahmoud A. Pouladi
- Subjects
Fragile X syndrome ,Human stem cells ,Protein synthesis ,Therapy ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract FXS is the most common genetic cause of intellectual (ID) and autism spectrum disorders (ASD). FXS is caused by loss of FMRP, an RNA-binding protein involved in the translational regulation of a large number of neuronal mRNAs. Absence of FMRP has been shown to lead to elevated protein synthesis and is thought to be a major cause of the synaptic plasticity and behavioural deficits in FXS. The increase in protein synthesis results in part from abnormal activation of key protein translation pathways downstream of ERK1/2 and mTOR signalling. Pharmacological and genetic interventions that attenuate hyperactivation of these pathways can normalize levels of protein synthesis and improve phenotypic outcomes in animal models of FXS. Several efforts are currently underway to trial this strategy in patients with FXS. To date, elevated global protein synthesis as a result of FMRP loss has not been validated in the context of human neurons. Here, using an isogenic human stem cell-based model, we show that de novo protein synthesis is elevated in FMRP-deficient neural cells. We further show that this increase is associated with elevated ERK1/2 and Akt signalling and can be rescued by metformin treatment. Finally, we examined the effect of normalizing protein synthesis on phenotypic abnormalities in FMRP-deficient neural cells. We find that treatment with metformin attenuates the increase in proliferation of FMRP-deficient neural progenitor cells but not the neuronal deficits in neurite outgrowth. The elevated level of protein synthesis and the normalization of neural progenitor proliferation by metformin treatment were validated in additional control and FXS patient-derived hiPSC lines. Overall, our results validate that loss of FMRP results in elevated de novo protein synthesis in human neurons and suggest that approaches targeting this abnormality are likely to be of partial therapeutic benefit in FXS.
- Published
- 2020
- Full Text
- View/download PDF
8. Loss-of-function mutations in UDP-Glucose 6-Dehydrogenase cause recessive developmental epileptic encephalopathy
- Author
-
Holger Hengel, Célia Bosso-Lefèvre, George Grady, Emmanuelle Szenker-Ravi, Hankun Li, Sarah Pierce, Élise Lebigot, Thong-Teck Tan, Michelle Y. Eio, Gunaseelan Narayanan, Kagistia Hana Utami, Monica Yau, Nader Handal, Werner Deigendesch, Reinhard Keimer, Hiyam M. Marzouqa, Meral Gunay-Aygun, Michael J. Muriello, Helene Verhelst, Sarah Weckhuysen, Sonal Mahida, Sakkubai Naidu, Terrence G. Thomas, Jiin Ying Lim, Ee Shien Tan, Damien Haye, Michèl A. A. P. Willemsen, Renske Oegema, Wendy G. Mitchell, Tyler Mark Pierson, Marisa V. Andrews, Marcia C. Willing, Lance H. Rodan, Tahsin Stefan Barakat, Marjon van Slegtenhorst, Ralitza H. Gavrilova, Diego Martinelli, Tal Gilboa, Abdullah M. Tamim, Mais O. Hashem, Moeenaldeen D. AlSayed, Maha M. Abdulrahim, Mohammed Al-Owain, Ali Awaji, Adel A. H. Mahmoud, Eissa A. Faqeih, Ali Al Asmari, Sulwan M. Algain, Lamyaa A. Jad, Hesham M. Aldhalaan, Ingo Helbig, David A. Koolen, Angelika Riess, Ingeborg Kraegeloh-Mann, Peter Bauer, Suleyman Gulsuner, Hannah Stamberger, Alvin Yu Jin Ng, Sha Tang, Sumanty Tohari, Boris Keren, Laura E. Schultz-Rogers, Eric W. Klee, Sabina Barresi, Marco Tartaglia, Hagar Mor-Shaked, Sateesh Maddirevula, Amber Begtrup, Aida Telegrafi, Rolph Pfundt, Rebecca Schüle, Brian Ciruna, Carine Bonnard, Mahmoud A. Pouladi, James C. Stewart, Adam Claridge-Chang, Dirk J. Lefeber, Fowzan S. Alkuraya, Ajay S. Mathuru, Byrappa Venkatesh, Joseph J. Barycki, Melanie A. Simpson, Saumya S. Jamuar, Ludger Schöls, and Bruno Reversade
- Subjects
Science - Abstract
UDP-glucuronic acid is a component of the extracellular matrix. Here, the authors report biallelic variants in the gene encoding UDP-Glucose 6-Dehydrogenase (UGDH) in individuals affected by developmental epileptic encephalopathies that impair UGDH stability, oligomerization, or enzymatic activity in vitro.
- Published
- 2020
- Full Text
- View/download PDF
9. Rescue of aberrant huntingtin palmitoylation ameliorates mutant huntingtin-induced toxicity
- Author
-
Fanny L. Lemarié, Nicholas S. Caron, Shaun S. Sanders, Mandi E. Schmidt, Yen T.N. Nguyen, Seunghyun Ko, Xiaohong Xu, Mahmoud A. Pouladi, Dale D.O. Martin, and Michael R. Hayden
- Subjects
Palmitoylation ,S-acylation ,Huntingtin ,Huntington disease ,ZDHHC17 ,Acyl-protein thioesterase ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Huntington disease (HD) is a neurodegenerative disorder caused by a CAG expansion in the HTT gene that codes for an elongated polyglutamine tract in the huntingtin (HTT) protein. HTT is subject to multiple post-translational modifications (PTMs) that regulate its cellular function. Mutating specific PTM sites within mutant HTT (mHTT) in HD mouse models can modulate disease phenotypes, highlighting the key role of HTT PTMs in the pathogenesis of HD. These findings have led to increased interest in developing small molecules to modulate HTT PTMs in order to decrease mHTT toxicity. However, the therapeutic efficacy of pharmacological modulation of HTT PTMs in preclinical HD models remains largely unknown. HTT is palmitoylated at cysteine 214 by the huntingtin-interacting protein 14 (HIP14 or ZDHHC17) and 14-like (HIP14L or ZDHHC13) acyltransferases. Here, we assessed if HTT palmitoylation should be regarded as a therapeutic target to treat HD by (1) investigating palmitoylation dysregulation in rodent and human HD model systems, (2) measuring the impact of mHTT-lowering therapy on brain palmitoylation, and (3) evaluating if HTT palmitoylation can be pharmacologically modulated. We show that palmitoylation of mHTT and some HIP14/HIP14L-substrates is decreased early in multiple HD mouse models, and that mHTT palmitoylation decreases further with aging. Lowering mHTT in the brain of YAC128 mice is not sufficient to rescue aberrant palmitoylation. However, we demonstrate that mHTT palmitoylation can be normalized in COS-7 cells, in YAC128 cortico-striatal primary neurons and HD patient-derived lymphoblasts using an acyl-protein thioesterase (APT) inhibitor. Moreover, we show that modulating palmitoylation reduces mHTT aggregation and mHTT-induced cytotoxicity in COS-7 cells and YAC128 neurons.
- Published
- 2021
- Full Text
- View/download PDF
10. Evidences for Mutant Huntingtin Inducing Musculoskeletal and Brain Growth Impairments via Disturbing Testosterone Biosynthesis in Male Huntington Disease Animals
- Author
-
Libo Yu-Taeger, Arianna Novati, Jonasz Jeremiasz Weber, Elisabeth Singer-Mikosch, Ann-Sophie Pabst, Fubo Cheng, Carsten Saft, Jennifer Koenig, Gisa Ellrichmann, Taneli Heikkinen, Mahmoud A. Pouladi, Olaf Riess, and Huu Phuc Nguyen
- Subjects
Huntington disease ,musculoskeletal growth ,brain growth ,sex-difference ,BACHD rats ,R6/2 mice ,Cytology ,QH573-671 - Abstract
Body weight (BW) loss and reduced body mass index (BMI) are the most common peripheral alterations in Huntington disease (HD) and have been found in HD mutation carriers and HD animal models before the manifestation of neurological symptoms. This suggests that, at least in the early disease stage, these changes could be due to abnormal tissue growth rather than tissue atrophy. Moreover, BW and BMI are reported to be more affected in males than females in HD animal models and patients. Here, we confirmed sex-dependent growth alterations in the BACHD rat model for HD and investigated the associated contributing factors. Our results showed growth abnormalities along with decreased plasma testosterone and insulin-like growth factor 1 (IGF-1) levels only in males. Moreover, we demonstrated correlations between growth parameters, IGF-1, and testosterone. Our analyses further revealed an aberrant transcription of testosterone biosynthesis-related genes in the testes of BACHD rats with undisturbed luteinizing hormone (LH)/cAMP/PKA signaling, which plays a key role in regulating the transcription process of some of these genes. In line with the findings in BACHD rats, analyses in the R6/2 mouse model of HD showed similar results. Our findings support the view that mutant huntingtin may induce abnormal growth in males via the dysregulation of gene transcription in the testis, which in turn can affect testosterone biosynthesis.
- Published
- 2022
- Full Text
- View/download PDF
11. Manipulation of microbiota reveals altered callosal myelination and white matter plasticity in a model of Huntington disease
- Author
-
Carola I. Radulescu, Marta Garcia-Miralles, Harwin Sidik, Costanza Ferrari Bardile, Nur Amirah Binte Mohammad Yusof, Hae Ung Lee, Eliza Xin Pei Ho, Collins Wenhan Chu, Emma Layton, Donovan Low, Paola Florez De Sessions, Sven Pettersson, Florent Ginhoux, and Mahmoud A. Pouladi
- Subjects
Huntington disease ,BACHD ,White matter ,Myelination ,Oligodendrocytes ,Germ-free ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Structural and molecular myelination deficits represent early pathological features of Huntington disease (HD). Recent evidence from germ-free (GF) animals suggests a role for microbiota-gut-brain bidirectional communication in the regulation of myelination. In this study, we aimed to investigate the impact of microbiota on myelin plasticity and oligodendroglial population dynamics in the mixed-sex BACHD mouse model of HD. Ultrastructural analysis of myelin in the corpus callosum revealed alterations of myelin thickness in BACHD GF compared to specific-pathogen free (SPF) mice, whereas no differences were observed between wild-type (WT) groups. In contrast, myelin compaction was altered in all groups when compared to WT SPF animals. Levels of myelin-related proteins were generally reduced, and the number of mature oligodendrocytes was decreased in the prefrontal cortex under GF compared to SPF conditions, regardless of genotype. Minor differences in commensal bacteria at the family and genera levels were found in the gut microbiota of BACHD and WT animals housed in standard living conditions. Our findings indicate complex effects of a germ-free status on myelin-related characteristics, and highlight the adaptive properties of myelination as a result of environmental manipulation.
- Published
- 2019
- Full Text
- View/download PDF
12. Reprint of: Manipulation of microbiota reveals altered callosal myelination and white matter plasticity in a model of Huntington disease
- Author
-
Carola I. Radulescu, Marta Garcia-Miralles, Harwin Sidik, Costanza Ferrari Bardile, Nur Amirah Binte Mohammad Yusof, Hae Ung Lee, Eliza Xin Pei Ho, Collins Wenhan Chu, Emma Layton, Donovan Low, Paola Florez De Sessions, Sven Pettersson, Florent Ginhoux, and Mahmoud A. Pouladi
- Subjects
Huntington disease ,BACHD ,White matter ,Myelination ,Oligodendrocytes ,Germ-free ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Structural and molecular myelination deficits represent early pathological features of Huntington disease (HD). Recent evidence from germ-free (GF) animals suggests a role for microbiota-gut-brain bidirectional communication in the regulation of myelination. In this study, we aimed to investigate the impact of microbiota on myelin plasticity and oligodendroglial population dynamics in the mixed-sex BACHD mouse model of HD. Ultrastructural analysis of myelin in the corpus callosum revealed alterations of myelin thickness in BACHD GF compared to specific-pathogen free (SPF) mice, whereas no differences were observed between wild-type (WT) groups. In contrast, myelin compaction was altered in all groups when compared to WT SPF animals. Levels of myelin-related proteins were generally reduced, and the number of mature oligodendrocytes was decreased in the prefrontal cortex under GF compared to SPF conditions, regardless of genotype. Minor differences in commensal bacteria at the family and genera levels were found in the gut microbiota of BACHD and WT animals housed in standard living conditions. Our findings indicate complex effects of a germ-free status on myelin-related characteristics, and highlight the adaptive properties of myelination as a result of environmental manipulation.
- Published
- 2020
- Full Text
- View/download PDF
13. A thiol probe for measuring unfolded protein load and proteostasis in cells
- Author
-
Moore Z. Chen, Nagaraj S. Moily, Jessica L. Bridgford, Rebecca J. Wood, Mona Radwan, Trevor A. Smith, Zhegang Song, Ben Zhong Tang, Leann Tilley, Xiaohong Xu, Gavin E. Reid, Mahmoud A. Pouladi, Yuning Hong, and Danny M. Hatters
- Subjects
Science - Abstract
Proteostasis is maintained through a number of molecular mechanisms, some of which function to protect the folded state of proteins. Here the authors demonstrate the use of TPE-MI in a fluorigenic dye assay for the quantitation of unfolded proteins that can be used to assess proteostasis on a cellular or proteome scale.
- Published
- 2017
- Full Text
- View/download PDF
14. Reversal of Phenotypic Abnormalities by CRISPR/Cas9-Mediated Gene Correction in Huntington Disease Patient-Derived Induced Pluripotent Stem Cells
- Author
-
Xiaohong Xu, Yilin Tay, Bernice Sim, Su-In Yoon, Yihui Huang, Jolene Ooi, Kagistia Hana Utami, Amin Ziaei, Bryan Ng, Carola Radulescu, Donovan Low, Alvin Yu Jin Ng, Marie Loh, Byrappa Venkatesh, Florent Ginhoux, George J. Augustine, and Mahmoud A. Pouladi
- Subjects
human induced pluripotent stem cell (hiPSC) ,neurodegenerative disorders ,Huntington disease ,genetic editing ,disease modeling ,disease phenotypes ,transcriptional dysrgulation ,CHCHD2 ,mitochondrial dysfunction ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Huntington disease (HD) is a dominant neurodegenerative disorder caused by a CAG repeat expansion in HTT. Here we report correction of HD human induced pluripotent stem cells (hiPSCs) using a CRISPR-Cas9 and piggyBac transposon-based approach. We show that both HD and corrected isogenic hiPSCs can be differentiated into excitable, synaptically active forebrain neurons. We further demonstrate that phenotypic abnormalities in HD hiPSC-derived neural cells, including impaired neural rosette formation, increased susceptibility to growth factor withdrawal, and deficits in mitochondrial respiration, are rescued in isogenic controls. Importantly, using genome-wide expression analysis, we show that a number of apparent gene expression differences detected between HD and non-related healthy control lines are absent between HD and corrected lines, suggesting that these differences are likely related to genetic background rather than HD-specific effects. Our study demonstrates correction of HD hiPSCs and associated phenotypic abnormalities, and the importance of isogenic controls for disease modeling using hiPSCs.
- Published
- 2017
- Full Text
- View/download PDF
15. Generation of four H1 hESC sublines carrying a hemizygous knock-out/mutant MECP2
- Author
-
Ruizhu Zeng, Harwin Sidik, Kim S. Robinson, Franklin L. Zhong, Bruno Reversade, and Mahmoud A. Pouladi
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Rett syndrome (RTT) is a childhood neurodevelopmental disorder caused by mutations in MECP2. To study the molecular mechanisms underlying RTT, four sublines of H1 hESCs were generated, carrying a hemizygous knockout or mutant allele of MECP2. Exons 3 and 4 of MECP2 were targeted using the CRISPR/Cas9 nuclease system.
- Published
- 2019
- Full Text
- View/download PDF
16. Generation of the Human Pluripotent Stem-Cell-Derived Astrocyte Model with Forebrain Identity
- Author
-
Ulla-Kaisa Peteri, Juho Pitkonen, Kagistia Hana Utami, Jere Paavola, Laurent Roybon, Mahmoud A. Pouladi, and Maija L. Castrén
- Subjects
pluripotent stem cells ,differentiation ,astrocytes ,patterning ,fragile X syndrome ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Astrocytes form functionally and morphologically distinct populations of cells with brain-region-specific properties. Human pluripotent stem cells (hPSCs) offer possibilities to generate astroglia for studies investigating mechanisms governing the emergence of astrocytic diversity. We established a method to generate human astrocytes from hPSCs with forebrain patterning and final specification with ciliary neurotrophic factor (CNTF). Transcriptome profiling and gene enrichment analysis monitored the sequential expression of genes determining astrocyte differentiation and confirmed activation of forebrain differentiation pathways at Day 30 (D30) and D60 of differentiation in vitro. More than 90% of astrocytes aged D95 in vitro co-expressed the astrocytic markers glial fibrillary acidic protein (GFAP) and S100β. Intracellular calcium responses to ATP indicated differentiation of the functional astrocyte population with constitutive monocyte chemoattractant protein-1 (MCP-1/CCL2) and tissue inhibitor of metalloproteinases-2 (TIMP-2) expression. The method was reproducible across several hPSC lines, and the data demonstrated the usefulness of forebrain astrocyte modeling in research investigating forebrain pathology.
- Published
- 2021
- Full Text
- View/download PDF
17. Anti-semaphorin 4D immunotherapy ameliorates neuropathology and some cognitive impairment in the YAC128 mouse model of Huntington disease
- Author
-
Amber L. Southwell, Sonia Franciosi, Erika B. Villanueva, Yuanyun Xie, Laurie A. Winter, Janaki Veeraraghavan, Alan Jonason, Boguslaw Felczak, Weining Zhang, Vlad Kovalik, Sabine Waltl, George Hall, Mahmoud A. Pouladi, Ernest S. Smith, William J. Bowers, Maurice Zauderer, and Michael R. Hayden
- Subjects
Semaphorin ,Immunotherapy ,Passive immunization ,Huntington disease ,Preclinical ,Transgenic mice ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Huntington disease (HD) is an inherited, fatal neurodegenerative disease with no disease-modifying therapy currently available. In addition to characteristic motor deficits and atrophy of the caudate nucleus, signature hallmarks of HD include behavioral abnormalities, immune activation, and cortical and white matter loss. The identification and validation of novel therapeutic targets that contribute to these degenerative cellular processes may lead to new interventions that slow or even halt the course of this insidious disease. Semaphorin 4D (SEMA4D) is a transmembrane signaling molecule that modulates a variety of processes central to neuroinflammation and neurodegeneration including glial cell activation, neuronal growth cone collapse and apoptosis of neural precursors, as well as inhibition of oligodendrocyte migration, differentiation and process formation. Therefore, inhibition of SEMA4D signaling could reduce CNS inflammation, increase neuronal outgrowth and enhance oligodendrocyte maturation, which may be of therapeutic benefit in the treatment of several neurodegenerative diseases, including HD. To that end, we evaluated the preclinical therapeutic efficacy of an anti-SEMA4D monoclonal antibody, which prevents the interaction between SEMA4D and its receptors, in the YAC128 transgenic HD mouse model. Anti-SEMA4D treatment ameliorated neuropathological signatures, including striatal atrophy, cortical atrophy, and corpus callosum atrophy and prevented testicular degeneration in YAC128 mice. In parallel, a subset of behavioral symptoms was improved in anti-SEMA4D treated YAC128 mice, including reduced anxiety-like behavior and rescue of cognitive deficits. There was, however, no discernible effect on motor deficits. The preservation of brain gray and white matter and improvement in behavioral measures in YAC128 mice treated with anti-SEMA4D suggest that this approach could represent a viable therapeutic strategy for the treatment of HD. Importantly, this work provides in vivo demonstration that inhibition of pathways initiated by SEMA4D constitutes a novel approach to moderation of neurodegeneration.
- Published
- 2015
- Full Text
- View/download PDF
18. Partial rescue of some features of Huntington Disease in the genetic absence of caspase-6 in YAC128 mice
- Author
-
Bibiana K.Y. Wong, Dagmar E. Ehrnhoefer, Rona K. Graham, Dale D.O. Martin, Safia Ladha, Valeria Uribe, Lisa M. Stanek, Sonia Franciosi, Xiaofan Qiu, Yu Deng, Vlad Kovalik, Weining Zhang, Mahmoud A. Pouladi, Lamya S. Shihabuddin, and Michael R. Hayden
- Subjects
Caspase-6 ,YAC128 ,Huntington Disease ,Autophagy ,p62 ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Huntington Disease (HD) is a progressive neurodegenerative disease caused by an elongated CAG repeat in the huntingtin (HTT) gene that encodes a polyglutamine tract in the HTT protein. Proteolysis of the mutant HTT protein (mHTT) has been detected in human and murine HD brains and is implicated in the pathogenesis of HD. Of particular importance is the site at amino acid (aa) 586 that contains a caspase-6 (Casp6) recognition motif.Activation of Casp6 occurs presymptomatically in human HD patients and the inhibition of mHTT proteolysis at aa586 in the YAC128 mouse model results in the full rescue of HD-like phenotypes. Surprisingly, Casp6 ablation in two different HD mouse models did not completely prevent the generation of this fragment, and therapeutic benefits were limited, questioning the role of Casp6 in the disease.We have evaluated the impact of the loss of Casp6 in the YAC128 mouse model of HD. Levels of the mHTT-586 fragment are reduced but not absent in the absence of Casp6 and we identify caspase 8 as an alternate enzyme that can generate this fragment. In vivo, the ablation of Casp6 results in a partial rescue of body weight gain, normalized IGF-1 levels, a reversal of the depression-like phenotype and decreased HTT levels. In the YAC128/Casp6−/− striatum there is a concomitant reduction in p62 levels, a marker of autophagic activity, suggesting increased autophagic clearance. These results implicate the HTT-586 fragment as a key contributor to certain features of HD, irrespective of the enzyme involved in its generation.
- Published
- 2015
- Full Text
- View/download PDF
19. NP03, a novel low-dose lithium formulation, is neuroprotective in the YAC128 mouse model of Huntington disease
- Author
-
Mahmoud A. Pouladi, Elsa Brillaud, Yuanyun Xie, Paola Conforti, Rona K. Graham, Dagmar E. Ehrnhoefer, Sonia Franciosi, Weining Zhang, Patrick Poucheret, Elsa Compte, Jean-Claude Maurel, Chiara Zuccato, Elena Cattaneo, Christian Néri, and Michael R. Hayden
- Subjects
Huntington disease ,Transgenic mouse model ,Lithium ,Caspase-6 ,BDNF ,GSK-3 ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Huntington disease (HD), a neurodegenerative disorder caused by an expanded CAG repeat in the HTT gene, remains without a treatment to modify the course of the illness. Lithium, a drug widely used for the treatment of bipolar disorder, has been shown to exert neuroprotective effects in a number of models of neurological disease but may have various toxic effects at conventional therapeutic doses. We examined whether NP03, a novel low-dose lithium microemulsion, would improve the disease phenotypes in the YAC128 mouse model of HD. We demonstrate that NP03 improves motor function, ameliorates the neuropathological deficits in striatal volume, neuronal counts, and DARPP-32 expression, and partially rescues testicular atrophy in YAC128 mice. These positive effects were accompanied by improvements in multiple biochemical endpoints associated with the pathogenesis of HD, including normalization of caspase-6 activation and amelioration of deficits in BDNF levels, and with no lithium-related toxicity. Our findings demonstrate that NP03 ameliorates the motor and neuropathological phenotypes in the YAC128 mouse model of HD, and represents a potential therapeutic approach for HD.
- Published
- 2012
- Full Text
- View/download PDF
20. Altered adult hippocampal neurogenesis in the YAC128 transgenic mouse model of Huntington disease
- Author
-
Jessica M. Simpson, Joana Gil-Mohapel, Mahmoud A. Pouladi, Mohamed Ghilan, Yuanyun Xie, Michael R. Hayden, and Brian R. Christie
- Subjects
Huntington disease ,Adult neurogenesis ,Cell proliferation ,Dentate gyrus ,Subventricular zone ,YAC128 transgenic mice ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Perturbations in neurogenesis in the adult brain have been implicated in impaired learning and memory. In the present study, we investigated which stages of the neurogenic process are affected in the transgenic YAC128 mouse model of Huntington disease (HD). Hippocampal neuronal proliferation was altered in the dentate gyrus (DG) of YAC128 mice as compared with wild-type (WT) littermate controls in early symptomatic to end-stage mice. In addition, we detected a significantly lower number of immature neurons in the DG of young, pre-symptomatic YAC128 mice. This decrease in neuronal differentiation persisted through the progression of the disease, and resulted in an overall reduction in the number of new mature neurons in the DG of YAC128 mice. There were no changes in cell proliferation and differentiation in the subventricular zone (SVZ). In this study, we demonstrate decreases in neurogenesis in the DG of YAC128 mice, and these deficits may contribute to the cognitive abnormalities observed in these animals.
- Published
- 2011
- Full Text
- View/download PDF
21. Maturation Delay of Human GABAergic Neurogenesis in Fragile X Syndrome Pluripotent Stem Cells
- Author
-
Ai Zhang, Irina Sokolova, Alain Domissy, Joshua Davis, Lee Rao, Kagistia Hana Utami, Yanling Wang, Randi J Hagerman, Mahmoud A Pouladi, Pietro Sanna, Michael J Boland, and Jeanne F Loring
- Subjects
Pluripotent Stem Cells ,Autism Spectrum Disorder ,Intellectual and Developmental Disabilities (IDD) ,Neurogenesis ,Medical Biotechnology ,Clinical Sciences ,autism ,Regenerative Medicine ,Epigenesis, Genetic ,Fragile X Mental Retardation Protein ,Rare Diseases ,Genetic ,Stem Cell Research - Nonembryonic - Human ,Genetics ,2.1 Biological and endogenous factors ,Humans ,fragile X syndrome ,Stem Cell Research - Embryonic - Human ,Aetiology ,Pediatric ,GABAergic neurons ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Neurosciences ,Cell Biology ,General Medicine ,Stem Cell Research ,Brain Disorders ,Mental Health ,hESCs ,Fragile X Syndrome ,Neurological ,Stem Cell Research - Nonembryonic - Non-Human ,human iPSCs ,Biochemistry and Cell Biology ,Epigenesis ,Developmental Biology - Abstract
Fragile X Syndrome (FXS), the leading monogenic cause of intellectual disability and autism spectrum disorder, is caused by expansion of a CGG trinucleotide repeat in the 5ʹ-UTR of the Fragile X Mental Retardation-1 (FMR1) gene. Epigenetic silencing of FMR1 results in loss of the Fragile X Mental Retardation Protein (FMRP). Although most studies to date have focused on excitatory neurons, recent evidence suggests that GABAergic inhibitory networks are also affected. To investigate human GABAergic neurogenesis, we established a method to reproducibly derive inhibitory neurons from multiple FXS and control human pluripotent stem cell (hPSC) lines. Electrophysiological analyses suggested that the developing FXS neurons had a delay in the GABA functional switch, a transition in fetal development that converts the GABAA channel’s function from depolarization to hyperpolarization, with profound effects on the developing brain. To investigate the cause of this delay, we analyzed 14 400 single-cell transcriptomes from FXS and control cells at 2 stages of GABAergic neurogenesis. While control and FXS cells were similar at the earlier time point, the later-stage FXS cells retained expression of neuroblast proliferation-associated genes and had lower levels of genes associated with action potential regulation, synapses, and mitochondria compared with controls. Our analysis suggests that loss of FMRP prolongs the proliferative stage of progenitors, which may result in more neurons remaining immature during the later stages of neurogenesis. This could have profound implications for homeostatic excitatory-inhibitory circuit development in FXS, and suggests a novel direction for understanding disease mechanisms that may help to guide therapeutic interventions.
- Published
- 2022
22. Widespread dysregulation of mRNA splicing implicates RNA processing in the development and progression of Huntington’s disease
- Author
-
Vincent Tano, Kagistia Hana Utami, Nur Amirah Binte Mohammad Yusof, Mahmoud A Pouladi, and Sarah R Langley
- Abstract
In Huntington’s disease (HD), a CAG repeat expansion mutation in theHTTgene drives a gain-of-function toxicity that disrupts mRNA processing. Although widespread dysregulation of gene splicing in the striatum has been shown in human HD post-mortem brain tissue, post-mortem analyses are likely confounded by cell type composition changes due to neuronal loss and astrogliosis in late stage HD. This limits the ability to identify dysregulation related to early pathogenesis. To study alternative splicing changes in early HD, we performed RNA-sequencing analysis in an established isogenic HD neuronal cell model. We report cell type-associated and CAG length-dependent splicing changes, and find an enrichment of RNA processing genes coupled with neuronal function-related genes showing mutantHTT-associated splicing changes. Comparison with post-mortem data also identified splicing events associated with early pathogenesis that persist to later stages of disease. In summary, our results highlight splicing dysregulation in RNA processing genes in early and late-stage HD, which may lead to disrupted neuronal function and neuropathology.
- Published
- 2022
23. A24 Mutant huntingtin impairs musculoskeletal and brain growth via disturbing testosterone biosynthesis in male huntington disease animals
- Author
-
Libo Yu-Taeger, Arianna Novati, Jonasz Jeremiasz Weber, Elisabeth Singer-Mikosch, Ann-Sophie Pabst, Carsten Saft, Jennifer König, Gisa Ellrichmann, Taneli Heikkinen, Mahmoud A Pouladi, Olaf Riess, and Huu Phuc Nguyen
- Published
- 2022
24. Pluripotent stem cell-derived models of neurological diseases reveal early transcriptional heterogeneity
- Author
-
Cynthia C. Hession, Walaa Oweis, Lisa M. Ellerby, Mahmoud A. Pouladi, Xiaohong Xu, Malka Nissim-Rafinia, Matan Sorek, Xian Adiconis, Neville E. Sanjana, Congyi Lu, Xi Shi, Moria Maman, Jen Q. Pan, Eran Meshorer, Sean Simmons, Shahar Simon, Joshua Z. Levin, and Feng Zhang
- Subjects
Adult ,Pluripotent Stem Cells ,lcsh:QH426-470 ,Smart-seq2 ,Stem cell model ,Disease ,Biology ,Models, Biological ,Genetic Heterogeneity ,Huntington's disease ,Gene expression ,scRNA-seq ,medicine ,Transcriptional regulation ,Humans ,Gene Regulatory Networks ,Genetic Predisposition to Disease ,Single cell ,RNA-Seq ,Progenitor cell ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Research ,Gene Expression Profiling ,Neurodegenerative diseases ,High-Throughput Nucleotide Sequencing ,medicine.disease ,Phenotype ,Human genetics ,lcsh:Genetics ,Gene Expression Regulation ,lcsh:Biology (General) ,Transcriptional heterogeneity ,Mutation ,Single-Cell Analysis ,Genetic Background ,Neuroscience ,Neurological diseases ,Huntington’s disease - Abstract
Background Many neurodegenerative diseases develop only later in life, when cells in the nervous system lose their structure or function. In many forms of neurodegenerative diseases, this late-onset phenomenon remains largely unexplained. Results Analyzing single-cell RNA sequencing from Alzheimer’s disease (AD) and Huntington’s disease (HD) patients, we find increased transcriptional heterogeneity in disease-state neurons. We hypothesize that transcriptional heterogeneity precedes neurodegenerative disease pathologies. To test this idea experimentally, we use juvenile forms (72Q; 180Q) of HD iPSCs, differentiate them into committed neuronal progenitors, and obtain single-cell expression profiles. We show a global increase in gene expression variability in HD. Autophagy genes become more stable, while energy and actin-related genes become more variable in the mutant cells. Knocking down several differentially variable genes results in increased aggregate formation, a pathology associated with HD. We further validate the increased transcriptional heterogeneity in CHD8+/− cells, a model for autism spectrum disorder. Conclusions Overall, our results suggest that although neurodegenerative diseases develop over time, transcriptional regulation imbalance is present already at very early developmental stages. Therefore, an intervention aimed at this early phenotype may be of high diagnostic value.
- Published
- 2021
25. Gene expression profiles complement the analysis of genomic modifiers of the clinical onset of Huntington disease
- Author
-
Nicholas S. Caron, William Casazza, Sara Mostafavi, Galen E.B. Wright, Mahmoud A. Pouladi, Jolene Ooi, Xiaohong Xu, Michael R. Hayden, Colin J. D. Ross, Lorenzo Casal, and Bernard Ng
- Subjects
AcademicSubjects/SCI01140 ,Adult ,Male ,DNA Repair ,Biology ,Polymorphism, Single Nucleotide ,Receptors, G-Protein-Coupled ,03 medical and health sciences ,0302 clinical medicine ,Huntington's disease ,Genetics ,medicine ,Humans ,Age of Onset ,Cognitive decline ,Molecular Biology ,Gene ,Genetics (clinical) ,Aged ,Mismatch Repair Endonuclease PMS2 ,030304 developmental biology ,Genetic association ,Huntingtin Protein ,0303 health sciences ,Endodeoxyribonucleases ,Genome ,Genomics ,General Medicine ,Middle Aged ,medicine.disease ,Multifunctional Enzymes ,Phenotype ,Gene expression profiling ,Exodeoxyribonucleases ,Huntington Disease ,Gene Expression Regulation ,Organ Specificity ,Female ,General Article ,Sulfatases ,Age of onset ,Transcriptome ,Trinucleotide Repeat Expansion ,Trinucleotide repeat expansion ,030217 neurology & neurosurgery ,Genome-Wide Association Study - Abstract
Huntington disease (HD) is a neurodegenerative disorder that is caused by a CAG repeat expansion in HTT. The length of this repeat, however, only explains a proportion of the variability in age of onset in patients. Genome-wide association studies have identified modifiers that contribute toward a proportion of the observed variance. By incorporating tissue-specific transcriptomic information with these results, additional modifiers can be identified. We performed a transcriptome-wide association study assessing heritable differences in genetically determined expression in diverse tissues, with genome-wide data from over 4000 patients. Functional validation of prioritized genes was undertaken in isogenic HD stem cells and patient brains. Enrichment analyses were performed with biologically relevant gene sets to identify the core pathways. HD-associated gene coexpression modules were assessed for associations with neurological phenotypes in an independent cohort and to guide drug repurposing analyses. Transcriptomic analyses identified genes that were associated with age of HD onset and displayed colocalization with gene expression signals in brain tissue (FAN1, GPR161, PMS2, SUMF2), with supporting evidence from functional experiments. This included genes involved in DNA repair, as well as novel-candidate modifier genes that have been associated with other neurological conditions. Further, cortical coexpression modules were also associated with cognitive decline and HD-related traits in a longitudinal cohort. In summary, the combination of population-scale gene expression information with HD patient genomic data identified novel modifier genes for the disorder. Further, these analyses expanded the pathways potentially involved in modifying HD onset and prioritized candidate therapeutics for future study., Graphical Abstract Graphical Abstract Schematic representation of methodology employed to identify transcriptomic modifiers of Huntington disease (HD). Transcriptome-wide association analyses facilitated (A) the identification of top transcriptomic HD onset modifier genes, (B) relevant biologically diverse gene sets that are enriched for this trait and (C) human cortical coexpression modules that are relevant for HD onset and related phenotypes in an aging cohort, along with signature matching of HD signals for drug repurposing.
- Published
- 2020
26. A13 Abnormal spinal cord myelination due to oligodendrocyte dysfunction in a model of huntington disease
- Author
-
Mahmoud A. Pouladi, Reynard Quek, Costanza Ferrari Bardile, Marta Garcia-Miralles, Harwin Sidik, and Nur Amirah Binte Mohammad Yusof
- Subjects
Pathology ,medicine.medical_specialty ,Huntingtin ,business.industry ,Disease ,Grey matter ,Spinal cord ,medicine.disease ,Oligodendrocyte ,White matter ,Myelin ,medicine.anatomical_structure ,Atrophy ,medicine ,business - Abstract
Background The contribution of grey matter (GM) and white matter (WM) degeneration to the progressive brain atrophy in Huntington Disease (HD) has been well studied. The pathology of the spinal cord in HD is comparatively less well documented. Aim Here we sought to investigate spinal cord pathology in a mouse model of HD, and in particular WM and myelination abnormalities. Method To determine whether GM and WM regions are atrophied in the spinal cord of 12 months old BACHD mice, we measured the area of GM and WM in the spinal cord by histological assessment. Electron microscopy was used to analyze myelin fibers in the cervical area of the spinal cord, where g-ratio of myelinated axons was used as a measure of myelin thickness. To investigate the impact of inactivation of mutant huntingtin (mHTT) in oligodendroglia on these measures, we used the previously described BACHDxNG2Cre (BN) mouse line where mHTT is specifically reduced in oligodendrocyte progenitor cells (OPC). Results We show that spinal GM and WM areas are significantly atrophied in BACHD mice compared to wild-type (WT) controls. We further demonstrate that specific reduction of mHTT in oligodendroglial cells rescues the atrophy of spinal cord WM, but not GM, as observed in BACHD mice. Inactivation of mHTT in oligodendroglia had no effect on the density of oligodendroglial cells but enhanced the expression of myelin-related proteins in the spinal cord. Conclusions Overall, our findings firstly demonstrate that the myelination abnormalities observed in brain WM structures in HD extend to the spinal cord, and secondly, they suggest that specific expression of mHTT in oligodendrocytes contributes to such abnormalities.
- Published
- 2021
27. Urokinase plasminogen activator mediates changes in human astrocytes modeling fragile X syndrome
- Author
-
Ulla Kaisa Peteri, Juho Pitkonen, Mahmoud A. Pouladi, Antti Vaheri, Laurent Roybon, Maija L. Castrén, Iryna M. Ethell, Plinio C. Casarotto, Kagistia Hana Utami, Tomas Strandin, Otso Nieminen, Pádraic Corcoran, Ilario de Toma, NeuroDevDiseaseModelling, Medicum, Department of Physiology, Department of Virology, and Viral Zoonosis Research Unit
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,MIGRATION ,Autism Spectrum Disorder ,Population ,Induced Pluripotent Stem Cells ,Tropomyosin receptor kinase B ,Biology ,3124 Neurology and psychiatry ,Extracellular matrix ,PATHWAY ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,0302 clinical medicine ,astrocyte ,medicine ,neuronal plasticity ,Animals ,Humans ,Receptor ,education ,urokinase plasminogen activator ,EPILEPSY ,030304 developmental biology ,0303 health sciences ,education.field_of_study ,WNT/BETA-CATENIN ,Voltage-dependent calcium channel ,RECEPTOR ,CENTRAL-NERVOUS-SYSTEM ,3112 Neurosciences ,MOUSE MODEL ,GENE ,Urokinase-Type Plasminogen Activator ,Cell biology ,medicine.anatomical_structure ,DIFFERENTIATION ,Neurology ,Astrocytes ,Fragile X Syndrome ,Neuron ,030217 neurology & neurosurgery ,Intracellular ,Astrocyte ,RESPONSES - Abstract
The function of astrocytes intertwines with the extracellular matrix, whose neuron and glial cell-derived components shape neuronal plasticity. Astrocyte abnormalities have been reported in the brain of the mouse model for fragile X syndrome (FXS), the most common cause of inherited intellectual disability, and a monogenic cause of autism spectrum disorder. We compared human FXS and control astrocytes generated from human induced pluripotent stem cells and we found increased expression of urokinase plasminogen activator (uPA), which modulates degradation of extracellular matrix. Several pathways associated with uPA and its receptor function were activated in FXS astrocytes. Levels of uPA were also increased in conditioned medium collected from FXS hiPSC-derived astrocyte cultures and correlated inversely with intracellular Ca2+ responses to activation of L-type voltage-gated calcium channels in human astrocytes. Increased uPA augmented neuronal phosphorylation of TrkB within the docking site for the phospholipase-Cγ1 (PLCγ1), indicating effects of uPA on neuronal plasticity. Gene expression changes during neuronal differentiation preceding astrogenesis likely contributed to properties of astrocytes with FXS-specific alterations that showed specificity by not affecting differentiation of adenosine triphosphate (ATP)-responsive astrocyte population. To conclude, our studies identified uPA as an important regulator of astrocyte function and demonstrated that increased uPA in human FXS astrocytes modulated astrocytic responses and neuronal plasticity.
- Published
- 2021
28. Abnormal Spinal Cord Myelination due to Oligodendrocyte Dysfunction in a Model of Huntington's Disease
- Author
-
Mahmoud A. Pouladi, Marta Garcia-Miralles, Reynard Quek, Harwin Sidik, Costanza Ferrari Bardile, and Nur Amirah Binte Mohammad Yusof
- Subjects
Pathology ,medicine.medical_specialty ,Huntingtin ,Grey matter ,White matter ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Myelin ,Mice ,0302 clinical medicine ,Atrophy ,Huntington's disease ,medicine ,Animals ,Myelin Sheath ,030304 developmental biology ,0303 health sciences ,business.industry ,Spinal cord ,medicine.disease ,White Matter ,Oligodendrocyte ,Oligodendroglia ,medicine.anatomical_structure ,Huntington Disease ,Spinal Cord ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Abstract
Background: The relative contribution of grey matter (GM) and white matter (WM) degeneration to the progressive brain atrophy in Huntington’s disease (HD) has been well studied. The pathology of the spinal cord in HD is comparatively less well documented. Objective: We aim to characterize spinal cord WM abnormalities in a mouse model of HD and evaluate whether selective removal of mutant huntingtin (mHTT) from oligodendroglia rescues these deficits. Methods: Histological assessments were used to determine the area of GM and WM in the spinal cord of 12-month-old BACHD mice, while electron microscopy was used to analyze myelin fibers in the cervical area of the spinal cord. To investigate the impact of inactivation of mHTT in oligodendroglia on these measures, we used the previously described BACHDxNG2Cre mouse line where mHTT is specifically reduced in oligodendrocyte progenitor cells. Results: We show that spinal GM and WM areas are significantly atrophied in HD mice compared to wild-type controls. We further demonstrate that specific reduction of mHTT in oligodendroglial cells rescues the atrophy of spinal cord WM, but not GM, observed in HD mice. Inactivation of mHTT in oligodendroglia had no effect on the density of oligodendroglial cells but enhanced the expression of myelin-related proteins in the spinal cord. Conclusion: Our findings demonstrate that the myelination abnormalities observed in brain WM structures in HD extend to the spinal cord and suggest that specific expression of mHTT in oligodendrocytes contributes to such abnormalities.
- Published
- 2021
29. Manipulation of microbiota reveals altered callosal myelination and white matter plasticity in a model of Huntington disease
- Author
-
Costanza Ferrari Bardile, Florent Ginhoux, Nur Amirah Binte Mohammad Yusof, Emma Layton, Collins Wenhan Chu, Donovan Low, Sven Pettersson, Hae Ung Lee, Mahmoud A. Pouladi, Eliza Xin Pei Ho, Carola I. Radulescu, Harwin Sidik, Marta Garcia-Miralles, Paola Florez de Sessions, Lee Kong Chian School of Medicine (LKCMedicine), and Singapore Centre for Environmental Life Sciences and Engineering (SCELSE)
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Population ,Gut flora ,Corpus callosum ,digestive system ,Corpus Callosum ,lcsh:RC321-571 ,White matter ,Mice ,Myelination ,03 medical and health sciences ,Myelin ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,BACHD ,Medicine [Science] ,Microbiome ,Prefrontal cortex ,education ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Pathological ,Myelin Sheath ,education.field_of_study ,Neuronal Plasticity ,Germ-free ,biology ,Oligodendrocytes ,Huntington disease ,biology.organism_classification ,White Matter ,Gastrointestinal Microbiome ,Disease Models, Animal ,Huntington Disease ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,nervous system ,Neurology ,030217 neurology & neurosurgery - Abstract
Structural and molecular myelination deficits represent early pathological features of Huntington disease (HD). Recent evidence from germ-free (GF) animals suggests a role for microbiota-gut-brain bidirectional communication in the regulation of myelination. In this study, we aimed to investigate the impact of microbiota on myelin plasticity and oligodendroglial population dynamics in the mixed-sex BACHD mouse model of HD. Ultrastructural analysis of myelin in the corpus callosum revealed alterations of myelin thickness in BACHD GF compared to specific-pathogen free (SPF) mice, whereas no differences were observed between wild-type (WT) groups. In contrast, myelin compaction was altered in all groups when compared to WT SPF animals. Levels of myelin-related proteins were generally reduced, and the number of mature oligodendrocytes was decreased in the prefrontal cortex under GF compared to SPF conditions, regardless of genotype. Minor differences in commensal bacteria at the family and genera levels were found in the gut microbiota of BACHD and WT animals housed in standard living conditions. Our findings indicate complex effects of a germ-free status on myelin-related characteristics, and highlight the adaptive properties of myelination as a result of environmental manipulation. Agency for Science, Technology and Research (A*STAR) Nanyang Technological University Published version M.A.P. was supported by funds from theAgency for Science Technology and Research and the NationalUniversity of Singapore. S.P. was supported by funds from LKC Schoolof Medicine and SCELSE.
- Published
- 2019
30. Glutaminase deficiency caused by short tandem repeat expansion in GLS
- Author
-
Farhad Karbassi, Joke J.F.A. van Vugt, Daman Kumari, Doreen Dobritzsch, Britt I. Drögemöller, Michael A. Eberle, Clara D. M. van Karnebeek, Maja Tarailo-Graovac, Brett Trost, Ronald J. A. Wanders, Saikat Santra, Michel van Weeghel, Youdong Wang, Wyeth W. Wasserman, Marjolein Turkenburg, Xiao-Yan Wen, Andre B. P. van Kuilenburg, Meaghan J Jones, Jagdeep S. Walia, Koroboshka Brand-Arzamendi, Rene Leen, Julia L Macisaac, Hans R. Waterham, Laura A. Tseng, Michael S. Kobor, Charlotte Nguyen, Karen Usdin, Janet Koster, Indhu-Shree Rajan-Babu, Xiaohong Xu, Bernice Sim, Jinqiu Zhang, Jan H. Veldink, Meng Li, Egor Dolzhenko, Ryan K. C. Yuen, Stephen W. Scherer, Cassandra L. McDonald, Judith Meijer, Phillip A. Richmond, Bruce E. Hayward, C. J. Ross, Galen E.B. Wright, Mahmoud A. Pouladi, Michael T. Geraghty, AGEM - Amsterdam Gastroenterology Endocrinology Metabolism, Laboratory Genetic Metabolic Diseases, AGEM - Inborn errors of metabolism, Oncogenomics, ARD - Amsterdam Reproduction and Development, APH - Methodology, Paediatric Metabolic Diseases, ACS - Diabetes & metabolism, Laboratory Medicine, and Pediatric surgery
- Subjects
Untranslated region ,Male ,Ataxia ,Genotype ,Developmental Disabilities ,Glutamine ,030204 cardiovascular system & hematology ,Polymerase Chain Reaction ,Article ,03 medical and health sciences ,0302 clinical medicine ,Glutaminase ,Cerebellum ,medicine ,Humans ,030212 general & internal medicine ,Allele ,Gene ,Amino Acid Metabolism, Inborn Errors ,Exome sequencing ,Genetics ,Whole genome sequencing ,Whole Genome Sequencing ,business.industry ,General Medicine ,medicine.disease ,Phenotype ,Inborn error of metabolism ,Child, Preschool ,Mutation ,Female ,medicine.symptom ,Atrophy ,business ,Microsatellite Repeats - Abstract
We report an inborn error of metabolism caused by an expansion of a GCA-repeat tract in the 5′ untranslated region of the gene encoding glutaminase (GLS) that was identified through detailed clinical and biochemical phenotyping, combined with whole-genome sequencing. The expansion was observed in three unrelated patients who presented with an early-onset delay in overall development, progressive ataxia, and elevated levels of glutamine. In addition to ataxia, one patient also showed cerebellar atrophy. The expansion was associated with a relative deficiency of GLS messenger RNA transcribed from the expanded allele, which probably resulted from repeat-mediated chromatin changes upstream of the GLS repeat. Our discovery underscores the importance of careful examination of regions of the genome that are typically excluded from or poorly captured by exome sequencing.
- Published
- 2019
31. Rescue of aberrant huntingtin palmitoylation ameliorates mutant huntingtin-induced toxicity
- Author
-
Dale D.O. Martin, Yen T. Nguyen, Mandi E. Schmidt, Fanny Lemarié, Michael R. Hayden, Mahmoud A. Pouladi, Shaun S. Sanders, Nicholas S. Caron, Seunghyun Ko, and Xiaohong Xu
- Subjects
Male ,Huntingtin ,Mutant ,Pathogenesis ,Mice ,0302 clinical medicine ,Chlorocebus aethiops ,Lymphocytes ,Enzyme Inhibitors ,Cytotoxicity ,Neurons ,Huntingtin Protein ,0303 health sciences ,Chemistry ,Lymphoblast ,S-acylation ,Huntington disease ,Polyglutamine tract ,3. Good health ,Cell biology ,ZDHHC17 ,Neurology ,COS Cells ,Female ,lipids (amino acids, peptides, and proteins) ,Palmitoylation ,RC321-571 ,congenital, hereditary, and neonatal diseases and abnormalities ,Lipoylation ,Transgene ,Nerve Tissue Proteins ,Neurosciences. Biological psychiatry. Neuropsychiatry ,Biology ,Neuroprotection ,Cell Line ,03 medical and health sciences ,mental disorders ,Animals ,Humans ,Cysteine ,Adaptor Proteins, Signal Transducing ,030304 developmental biology ,Acyl-protein thioesterase ,Rats ,nervous system diseases ,nervous system ,Mutation ,Function (biology) ,Acyltransferases ,030217 neurology & neurosurgery - Abstract
Huntington disease (HD) is a neurodegenerative disorder caused by a CAG expansion in the HTT gene that codes for an elongated polyglutamine tract in the huntingtin (HTT) protein. HTT is subject to multiple post-translational modifications (PTMs) that regulate its cellular function. Mutating specific PTM sites within mutant HTT (mHTT) in HD mouse models can modulate disease phenotypes, highlighting the key role of HTT PTMs in the pathogenesis of HD. These findings have led to increased interest in developing small molecules to modulate HTT PTMs in order to decrease mHTT toxicity. However, the therapeutic efficacy of pharmacological modulation of HTT PTMs in preclinical HD models remains largely unknown. HTT is palmitoylated at cysteine 214 by the huntingtin-interacting protein 14 (HIP14 or ZDHHC17) and 14-like (HIP14L or ZDHHC13) acyltransferases. Here, we assessed if HTT palmitoylation should be regarded as a therapeutic target to treat HD by (1) investigating palmitoylation dysregulation in rodent and human HD model systems, (2) measuring the impact of mHTT-lowering therapy on brain palmitoylation, and (3) evaluating if HTT palmitoylation can be pharmacologically modulate. We show that palmitoylation of mHTT and some HIP14/HIP14L-substrates is decreased early in multiple HD mouse models, and that aging further reduces HTT palmitoylation. Lowering mHTT in the brain of YAC128 mice is not sufficient to rescue aberrant palmitoylation. However, we demonstrate that mHTT palmitoylation can be normalized in COS-7 cells, in YAC128 cortico-striatal primary neurons and HD patient-derived lymphoblasts using an acyl-protein thioesterase (APT) inhibitor. Moreover, we show that modulating palmitoylation reduces mHTT aggregation and mHTT-induced cytotoxicity in COS-7 cells and YAC128 neurons.HighlightsPalmitoylation of mHTT is reduced in multiple transgenic HD mouse modelsHTT palmitoylation decreases with increasing polyQ length in HD patient cellsmHTT-lowering in mouse brains does not rescue aberrant palmitoylationmHTT palmitoylation in HD patient-derived cells can be rescued via APT inhibitionPromoting palmitoylation reduces mHTT aggregation and cytotoxicity in vitro
- Published
- 2021
32. A Micropatterned Human-Specific Neuroepithelial Tissue for Modeling Gene and Drug-Induced Neurodevelopmental Defects
- Author
-
Bruno Reversade, Jean Jacques Clement Fatien, Jerome Zu Yao Tan, Mahmoud A. Pouladi, Geetika Sahni, Jeremy Teo Choon Meng, Yi-Chin Toh, Hülya Kayserili, Shu Yung Chang, Puck Wee Chan, Thong Teck Tan, Umut Altunoglu, Carine Bonnard, Kagistia Hana Utami, ACS - Heart failure & arrhythmias, Karabey, Hülya Kayserili (ORCID 0000-0003-0376-499X & YÖK ID 7945), Reversade, Bruno, Sahni, Geetika, Chang, Shu-Yung, Meng, Jeremy Teo Choon, Tan, Jerome Zu Yao, Fatien, Jean Jacques Clement, Bonnard, Carine, Utami, Kagistia Hana, Chan, Puck Wee, Tan, Thong Teck, Altunoglu, Umut, Pouladi, Mahmoud, Toh, Yi-Chin, and School of Medicine
- Subjects
Drug ,General Chemical Engineering ,media_common.quotation_subject ,Science ,Neuroepithelial Tissue ,General Physics and Astronomy ,Medicine (miscellaneous) ,morphogenesis ,Ectoderm ,02 engineering and technology ,Cell fate determination ,Biology ,010402 general chemistry ,Chemistry ,Nanoscience and nanotechnology ,Materials science ,01 natural sciences ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Corrections ,medicine ,General Materials Science ,human pluripotent stem cells ,Human specific ,lcsh:Science ,Gene ,media_common ,neurodevelopmental defects ,Full Paper ,Neural tube ,General Engineering ,Correction ,Apical constriction ,Full Papers ,neuroepithelium ,021001 nanoscience & nanotechnology ,Embryonic stem cell ,micropatterning ,0104 chemical sciences ,Cell biology ,Neuroepithelial cell ,medicine.anatomical_structure ,Neurulation ,Human pluripotent stem cells ,Micropatterning ,Morphogenesis ,Neurodevelopmental defects ,Neuroepithelium ,lcsh:Q ,0210 nano-technology ,Neuroscience - Abstract
The generation of structurally standardized human pluripotent stem cell (hPSC)‐derived neural embryonic tissues has the potential to model genetic and environmental mediators of early neurodevelopmental defects. Current neural patterning systems have so far focused on directing cell fate specification spatio‐temporally but not morphogenetic processes. Here, the formation of a structurally reproducible and highly‐organized neuroepithelium (NE) tissue is directed from hPSCs, which recapitulates morphogenetic cellular processes relevant to early neurulation. These include having a continuous, polarized epithelium and a distinct invagination‐like folding, where primitive ectodermal cells undergo E‐to‐N‐cadherin switching and apical constriction as they acquire a NE fate. This is accomplished by spatio‐temporal patterning of the mesoendoderm, which guides the development and self‐organization of the adjacent primitive ectoderm into the NE. It is uncovered that TGFβ signaling emanating from endodermal cells support tissue folding of the prospective NE. Evaluation of NE tissue structural dysmorphia, which is uniquely achievable in the model, enables the detection of apical constriction and cell adhesion dysfunctions in patient‐derived hPSCs as well as differentiating between different classes of neural tube defect‐inducing drugs., This paper reports the generation of a reproducible and highly organized neuroepithelial (NE) tissue by combining human pluripotent stem cell (hPSC) micropatterning and a temporally sequenced induction protocol to specify NE cells in spatial juxtaposition to mesoendoderm cells. By evaluating NE tissue structural dysmorphia in the micropatterned NE model, we can successfully model gene‐ and drug‐induced neurodevelopmental defects.
- Published
- 2021
33. Pluripotent stem cell derived models of neurological diseases reveal early transcriptional heterogeneity
- Author
-
Jen Q. Pan, Cynthia C. Hession, Walaa Oweis, Matan Sorek, Xian Adiconis, Malka Nissim-Rafinia, Neville E. Sanjana, Shahar Simon, Moria Maman, Lisa M. Ellerby, Congyi Lu, Joshua Z. Levin, Eran Meshorer, Xi Shi, Sean Simmons, Mahmoud A. Pouladi, Xiaohong Xu, and Feng Zhang
- Subjects
medicine.anatomical_structure ,Gene expression ,Cell ,medicine ,Transcriptional regulation ,Disease ,Biology ,Progenitor cell ,Induced pluripotent stem cell ,Neuroscience ,Gene ,Phenotype - Abstract
BackgroundMany neurodegenerative diseases (NDs) develop only later in life, when cells in the nervous system lose their structure or function. In genetic forms of NDs, this late onset phenomenon remains largely unexplained.ResultsAnalyzing single cell RNA sequencing (scRNA-seq) from Alzheimer’s disease (AD) patients, we find increased transcriptional heterogeneity in AD excitatory neurons. We hypothesized that transcriptional heterogeneity precedes ND pathologies. To test this idea experimentally, we used juvenile forms (72Q; 180Q) of Huntington’s disease (HD) iPSCs, differentiated them into committed neuronal progenitors, and obtained single cell expression profiles. We show a global increase in gene expression variability in HD. Autophagy genes become more stable, while energy and actin-related genes become more variable in the mutant cells. Knocking-down several differentially-variable genes resulted in increased aggregate formation, a pathology associated with HD. We further validated the increased transcriptional heterogeneity in CHD8+/- cells, a model for autism spectrum disorder.ConclusionsOverall, our results suggest that although NDs develop over time, transcriptional regulation imbalance is present already at very early developmental stages. Therefore, an intervention aimed at this early phenotype may be of high diagnostic value.
- Published
- 2020
34. Loss-of-function mutations in UDP-Glucose 6-Dehydrogenase cause recessive developmental epileptic encephalopathy
- Author
-
Ali Al Asmari, Emmanuelle Szenker-Ravi, Carine Bonnard, Bruno Reversade, Laura Schultz-Rogers, I. Kraegeloh-Mann, Maha Abdulrahim, Hesham Aldhalaan, Byrappa Venkatesh, Célia Bosso-Lefèvre, Aida Telegrafi, Hiyam M. Marzouqa, Gunaseelan Narayanan, Sha Tang, Sonal Mahida, Melanie A. Simpson, Fowzan S. Alkuraya, Michelle Eio, Eissa Faqeih, Renske Oegema, Sarah Weckhuysen, George Grady, Joseph J. Barycki, Mohammed Al-Owain, Lamyaa A. Jad, David A. Koolen, Marjon van Slegtenhorst, Tyler Mark Pierson, Marisa V. Andrews, Rebecca Schüle, Reinhard Keimer, Amber Begtrup, Sateesh Maddirevula, Michael Muriello, Sakkubai Naidu, Damien Haye, Adel A H Mahmoud, Brian Ciruna, Abdullah Tamim, Thong Teck Tan, Rolph Pfundt, Peter Bauer, Jiin Ying Lim, Ali Awaji, Marco Tartaglia, Meral Gunay-Aygun, Eric W. Klee, Marcia C. Willing, Monica Yau, Angelika Riess, Diego Martinelli, Sabina Barresi, Sumanty Tohari, Werner Deigendesch, Dirk Lefeber, Saumya Shekhar Jamuar, Ludger Schöls, Ralitza H. Gavrilova, Alvin Yu Jin Ng, Hannah Stamberger, Suleyman Gulsuner, Adam Claridge-Chang, Élise Lebigot, Moeenaldeen Al-Sayed, Ee Shien Tan, Kagistia Hana Utami, Sarah B. Pierce, Helene Verhelst, Hankun Li, James C. Stewart, Ingo Helbig, Tal Gilboa, Mahmoud A. Pouladi, Hagar Mor-Shaked, Boris Keren, Ajay S. Mathuru, Holger Hengel, Michèl A.A.P. Willemsen, Nader Handal, Tahsin Stefan Barakat, Sulwan M. Algain, Terrence Thomas, Lance H. Rodan, Mais Hashem, Wendy G. Mitchell, Center for Reproductive Medicine, ARD - Amsterdam Reproduction and Development, ACS - Diabetes & metabolism, Clinical Genetics, Reversade, Bruno, Hengel, H., Bosso-Lefèvre, C., Grady, G., Szenker-Ravi, E., Li, H., Pierce, S., Lebigot, É., Tan, T.-T., Eio, M.Y., Narayanan, G., Utami, K.H., Yau, M., Handal, N., Deigendesch, W., Keimer, R., Marzouqa, H.M., Gunay-Aygun, M., Muriello, M.J., Verhelst, H., Weckhuysen, S., Mahida, S., Naidu, S., Thomas, T.G., Lim, J.Y., Tan, E.S., Haye, D., Willemsen, M.A.A.P., Oegema, R., Mitchell, W.G., Pierson, T.M., Andrews, M.V., Willing, M.C., Rodan, L.H., Barakat, T.S., van Slegtenhorst, M., Gavrilova, R.H., Martinelli, D., Gilboa, T., Tamim, A.M., Hashem, M.O., AlSayed, M.D., Abdulrahim, M.M., Al-Owain, M., Awaji, A., Mahmoud, A.A.H., Faqeih, E.A., Asmari, A.A., Algain, S.M., Jad, L.A., Aldhalaan, H.M., Helbig, I., Koolen, D.A., Riess, A., Kraegeloh-Mann, I., Bauer, P., Gulsuner, S., Stamberger, H., Ng, A.Y.J., Tang, S., Tohari, S., Keren, B., Schultz-Rogers, L.E., Klee, E.W., Barresi, S., Tartaglia, M., Mor-Shaked, H., Maddirevula, S., Begtrup, A., Telegrafi, A., Pfundt, R., Schüle, R., Ciruna, B., Bonnard, C., Pouladi, M.A., Stewart, J.C., Claridge-Chang, A., Lefeber, D.J., Alkuraya, F.S., Mathuru, A.S., Venkatesh, B., Barycki, J.J., Simpson, M.A., Jamuar, S.S., Schöls, L, and School of Medicine
- Subjects
0301 basic medicine ,Male ,Glycobiology ,General Physics and Astronomy ,VARIANTS ,Encephalopathy ,Neurodegenerative ,Germline ,0302 clinical medicine ,UDP-GLUCOSE DEHYDROGENASE ,Loss of Function Mutation ,Medicine and Health Sciences ,EMBRYOGENESIS ,2.1 Biological and endogenous factors ,UGDH protein, human ,Aetiology ,Child ,lcsh:Science ,Zebrafish ,UTILITY ,Genetics ,pathology [Organoids] ,Multidisciplinary ,Uridine diphosphate glucose dehydrogenase ,Uridine diphosphate ,DP-glucuronic acid ,Syndrome ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,Hypotonia ,3. Good health ,Pedigree ,DEFICIENCY ,genetics [Loss of Function Mutation] ,Organoids ,genetics [Uridine Diphosphate Glucose Dehydrogenase] ,Child, Preschool ,Neurological ,Medicine ,Female ,ddc:500 ,medicine.symptom ,Oxidoreductases ,Engineering sciences. Technology ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,ENZYME ,Adolescent ,CONGENITAL DISORDER ,Science ,Intellectual and Developmental Disabilities (IDD) ,genetics [Epilepsy] ,chemistry [Oxidoreductases] ,Genetics and Molecular Biology ,Genes, Recessive ,Biology ,Uridine Diphosphate Glucose Dehydrogenase ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Protein Domains ,medicine ,Animals ,Humans ,Recessive ,Clinical genetics ,Allele ,Preschool ,Gene ,Loss function ,Alleles ,HEPARAN-SULFATE ,Phenocopy ,genetics [Oxidoreductases] ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,Epilepsy ,GLYCOSYLATION ,Neurosciences ,Infant ,General Chemistry ,biology.organism_classification ,medicine.disease ,Brain Disorders ,carbohydrates (lipids) ,Kinetics ,030104 developmental biology ,Genes ,General Biochemistry ,Neuronal development ,lcsh:Q ,Human medicine ,030217 neurology & neurosurgery ,Congenital disorder - Abstract
Developmental epileptic encephalopathies are devastating disorders characterized by intractable epileptic seizures and developmental delay. Here, we report an allelic series of germline recessive mutations in UGDH in 36 cases from 25 families presenting with epileptic encephalopathy with developmental delay and hypotonia. UGDH encodes an oxidoreductase that converts UDP-glucose to UDP-glucuronic acid, a key component of specific proteoglycans and glycolipids. Consistent with being loss-of-function alleles, we show using patients’ primary fibroblasts and biochemical assays, that these mutations either impair UGDH stability, oligomerization, or enzymatic activity. In vitro, patient-derived cerebral organoids are smaller with a reduced number of proliferating neuronal progenitors while mutant ugdh zebrafish do not phenocopy the human disease. Our study defines UGDH as a key player for the production of extracellular matrix components that are essential for human brain development. Based on the incidence of variants observed, UGDH mutations are likely to be a frequent cause of recessive epileptic encephalopathy., German Research Foundation (DFG); European Union (European Union); NEUROMICS Network; International Coordination Action (ICA); Fund for Scientific Research Flanders (FWO); Netherlands Organization for Scientific Research (ZONMW VIDI); National Medical Research Council, Singapore; A Strategic Positioning Fund on Genetic Orphan Diseases (GODAFIT); Industry Alignment Fund on Singapore Childhood Undiagnosed Diseases Program (SUREKids); Biomedical Research Council, A*STAR; Diana and Steve Marienhoff Fashion Industries Guild Endowed Fellowship in Pediatric Neuromuscular Diseases; Fondazione Bambino Gesù (Vite Coraggiose); Canadian Institutes of Health Research; Natural Sciences and Engineering Research Council of Canada; Eurocores Program EuroEPINOMICS; University of Antwerp Research Fund; FRAXA Foundation; Brain & Behavior Research Foundation, NARSAD Young Investigator Grant
- Published
- 2020
35. Integrative analysis identifies key molecular signatures underlying neurodevelopmental deficits in fragile X syndrome
- Author
-
Qiyu Chen, Marta Garcia-Miralles, Carola I. Radulescu, Bernice Sim, Xin Yi Yeo, Herty Liany, Georgia Chaldaiopoulou, Matthias Mann, Maija L. Castrén, Mahmoud A. Pouladi, Sangyong Jung, Srikanth Nama, Prabha Sampath, Han-Gyu Bae, Niels H. Skotte, Kagistia Hana Utami, Ana R. Colaço, Ulla-Kaisa Peteri, Nur Amirah Binte Mohammad Yusof, NeuroDevDiseaseModelling, Department of Physiology, Faculty of Medicine, and University of Helsinki
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,INTELLECTUAL DISABILITY ,Neurite ,Proteome ,Autism Spectrum Disorder ,Induced Pluripotent Stem Cells ,Context (language use) ,Biology ,MECHANISMS ,Transcriptome ,NEUROGENESIS ,03 medical and health sciences ,Fragile X Mental Retardation Protein ,0302 clinical medicine ,Neurodevelopmental disorder ,Human neurons ,medicine ,Humans ,Progenitor cell ,Induced pluripotent stem cell ,Biological Psychiatry ,Neurons ,ABNORMALITIES ,Neurogenesis ,Neurodevelopmental disorders ,3112 Neurosciences ,1184 Genetics, developmental biology, physiology ,MENTAL-RETARDATION PROTEIN ,MOUSE MODEL ,medicine.disease ,FMR1 ,Embryonic stem cell ,GENE ,nervous system diseases ,Fragile X syndrome ,SYNDROME NEURONS ,030104 developmental biology ,DIFFERENTIATION ,Ion transmembrane transporter activity ,Isogenic ,Neuron differentiation ,1182 Biochemistry, cell and molecular biology ,EMBRYONIC STEM-CELLS ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Background Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by epigenetic silencing of FMR1 and loss of FMRP expression. Efforts to understand the molecular underpinnings of the disease have been largely performed in rodent or nonisogenic settings. A detailed examination of the impact of FMRP loss on cellular processes and neuronal properties in the context of isogenic human neurons remains lacking. Methods Using CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 to introduce indels in exon 3 of FMR1, we generated an isogenic human pluripotent stem cell model of FXS that shows complete loss of FMRP expression. We generated neuronal cultures and performed genome-wide transcriptome and proteome profiling followed by functional validation of key dysregulated processes. We further analyzed neurodevelopmental and neuronal properties, including neurite length and neuronal activity, using multielectrode arrays and patch clamp electrophysiology. Results We showed that the transcriptome and proteome profiles of isogenic FMRP-deficient neurons demonstrate perturbations in synaptic transmission, neuron differentiation, cell proliferation and ion transmembrane transporter activity pathways, and autism spectrum disorder–associated gene sets. We uncovered key deficits in FMRP-deficient cells demonstrating abnormal neural rosette formation and neural progenitor cell proliferation. We further showed that FMRP-deficient neurons exhibit a number of additional phenotypic abnormalities, including neurite outgrowth and branching deficits and impaired electrophysiological network activity. These FMRP-deficient related impairments have also been validated in additional FXS patient–derived human-induced pluripotent stem cell neural cells. Conclusions Using isogenic human pluripotent stem cells as a model to investigate the pathophysiology of FXS in human neurons, we reveal key neural abnormalities arising from the loss of FMRP.
- Published
- 2020
36. Ermin deficiency as an inside-out model of inflammatory dysmyelination
- Author
-
Carola I. Radulescu, Alvin Yu Jin Ng, Bruno Reversade, Costanza Ferrari Bardile, Carine Bonnard, Harwin Sidik, Byrappa Venkatesh, Sarah R. Langley, Han-Gyu Bae, Amin Ziaei, Florent Ginhoux, Vahid Shaygannejad, Aymeric Silvin, Liang Juin Tan, Leila Dehghani, Mahmoud A. Pouladi, Sumanty Tohari, Marta Garcia-Miralles, Sangyong Jung, and Nur Amirah Binte Mohammad Yusof
- Subjects
Pathology ,medicine.medical_specialty ,Multiple sclerosis ,Central nervous system ,Saltatory conduction ,Biology ,medicine.disease ,Microgliosis ,Astrogliosis ,White matter ,Myelin ,medicine.anatomical_structure ,Knockout mouse ,medicine - Abstract
Ermin is an actin-binding protein found almost exclusively in the central nervous system (CNS) as a component of myelin sheaths. Although Ermin has been predicted to play a role in the formation and stability of myelin sheaths, this has not been directly examined in vivo. Here we show that Ermin is essential for myelin sheath integrity and normal saltatory conduction. Loss of Ermin in mice caused de-compacted and fragmented myelin sheaths and led to slower conduction along with progressive neurological deficits. RNA sequencing of the corpus callosum, the largest white matter structure in the CNS, pointed to inflammatory activation in aged Ermin-deficient mice, which was corroborated by increased levels of microgliosis and astrogliosis. The inflammatory milieu and myelin abnormalities were further associated with increased susceptibility to immune-mediated demyelination insult in Ermin knockout mice. Supporting a possible role of Ermin deficiency in inflammatory white matter disorders, a rare inactivating mutation in the ERMN gene was identified in multiple sclerosis patients. Our findings demonstrate a critical role for Ermin in maintaining myelin integrity. Given its near exclusive expression in myelinating oligodendrocytes, Ermin deficiency represents a compelling “inside-out” model of inflammatory dysmyelination and may offer a new paradigm for the development of myelin stability-targeted therapies.
- Published
- 2020
37. Elevated de novo protein synthesis in FMRP-deficient human neurons and its correction by metformin treatment
- Author
-
Nur Amirah Binte Mohammad Yusof, Maija L. Castrén, Mahmoud A. Pouladi, Kagistia Hana Utami, Jing Eugene Kwa, Ulla-Kaisa Peteri, NeuroDevDiseaseModelling, Medicum, Department of Physiology, Faculty of Medicine, and University of Helsinki
- Subjects
Human stem cells ,lcsh:RC346-429 ,3124 Neurology and psychiatry ,0302 clinical medicine ,Translational regulation ,Mitogen-Activated Protein Kinase 1 ,Neurons ,0303 health sciences ,Mitogen-Activated Protein Kinase 3 ,1184 Genetics, developmental biology, physiology ,PROLIFERATION ,MOUSE MODEL ,Neural stem cell ,Metformin ,3. Good health ,Cell biology ,Psychiatry and Mental health ,Stem cell ,STEM-CELLS ,medicine.drug ,congenital, hereditary, and neonatal diseases and abnormalities ,Neurite ,INHIBITION ,Context (language use) ,Biology ,Cell Line ,03 medical and health sciences ,Developmental Neuroscience ,medicine ,Humans ,FRAGILE-X-SYNDROME ,Molecular Biology ,lcsh:Neurology. Diseases of the nervous system ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,Cell Proliferation ,DYSREGULATION ,IDENTIFICATION ,Research ,Gene Expression Profiling ,3112 Neurosciences ,Fragile X Syndrome ,Protein Biosynthesis ,Synaptic plasticity ,Therapy ,Protein synthesis ,Proto-Oncogene Proteins c-akt ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
FXS is the most common genetic cause of intellectual (ID) and autism spectrum disorders (ASD). FXS is caused by loss of FMRP, an RNA-binding protein involved in the translational regulation of a large number of neuronal mRNAs. Absence of FMRP has been shown to lead to elevated protein synthesis and is thought to be a major cause of the synaptic plasticity and behavioural deficits in FXS. The increase in protein synthesis results in part from abnormal activation of key protein translation pathways downstream of ERK1/2 and mTOR signalling. Pharmacological and genetic interventions that attenuate hyperactivation of these pathways can normalize levels of protein synthesis and improve phenotypic outcomes in animal models of FXS. Several efforts are currently underway to trial this strategy in patients with FXS. To date, elevated global protein synthesis as a result of FMRP loss has not been validated in the context of human neurons. Here, using an isogenic human stem cell-based model, we show that de novo protein synthesis is elevated in FMRP-deficient neural cells. We further show that this increase is associated with elevated ERK1/2 and Akt signalling and can be rescued by metformin treatment. Finally, we examined the effect of normalizing protein synthesis on phenotypic abnormalities in FMRP-deficient neural cells. We find that treatment with metformin attenuates the increase in proliferation of FMRP-deficient neural progenitor cells but not the neuronal deficits in neurite outgrowth. The elevated level of protein synthesis and the normalization of neural progenitor proliferation by metformin treatment were validated in additional control and FXS patient-derived hiPSC lines. Overall, our results validate that loss of FMRP results in elevated de novo protein synthesis in human neurons and suggest that approaches targeting this abnormality are likely to be of partial therapeutic benefit in FXS.
- Published
- 2020
38. pS421 huntingtin modulates mitochondrial phenotypes and confers neuroprotection in an HD hiPSC model
- Author
-
Shuping Lin, Michael R. Hayden, Wah Ing Goh, Yoonjeong Cha, Chris Kay, Tamara Ratovitski, Bryan Tsong-Jye Ng, Rebecca Kusko, Carola I. Radulescu, Bernice Sim, Christopher A. Ross, John Soon Yew Lim, Nur Amirah Binte Mohammad Yusof, Graham D. Wright, Mahmoud A. Pouladi, and Xiaohong Xu
- Subjects
Cancer Research ,Huntingtin ,Immunology ,Mutant ,Cell death in the nervous system ,Context (language use) ,Oxidative phosphorylation ,Biology ,Neuroprotection ,Article ,Cellular and Molecular Neuroscience ,Mice ,Animals ,Humans ,lcsh:QH573-671 ,Induced pluripotent stem cell ,lcsh:Cytology ,Cell Biology ,Phenotype ,Cell biology ,Mitochondria ,Disease Models, Animal ,Induced pluripotent stem cells ,Huntington Disease ,Phosphorylation - Abstract
Huntington disease (HD) is a hereditary neurodegenerative disorder caused by mutant huntingtin (mHTT). Phosphorylation at serine-421 (pS421) of mHTT has been shown to be neuroprotective in cellular and rodent models. However, the genetic context of these models differs from that of HD patients. Here we employed human pluripotent stem cells (hiPSCs), which express endogenous full-length mHTT. Using genome editing, we generated isogenic hiPSC lines in which the S421 site in mHTT has been mutated into a phospho-mimetic aspartic acid (S421D) or phospho-resistant alanine (S421A). We observed that S421D, rather than S421A, confers neuroprotection in hiPSC-derived neural cells. Although we observed no effect of S421D on mHTT clearance or axonal transport, two aspects previously reported to be impacted by phosphorylation of mHTT at S421, our analysis revealed modulation of several aspects of mitochondrial form and function. These include mitochondrial surface area, volume, and counts, as well as improved mitochondrial membrane potential and oxidative phosphorylation. Our study validates the protective role of pS421 on mHTT and highlights a facet of the relationship between mHTT and mitochondrial changes in the context of human physiology with potential relevance to the pathogenesis of HD.
- Published
- 2020
39. Early deficits in olfaction are associated with structural and molecular alterations in the olfactory system of a Huntington disease mouse model
- Author
-
Mahmoud A. Pouladi, Blair R. Leavitt, Mélissa Laroche, E Peachey, Marta Garcia-Miralles, Mélissa Lessard-Beaudoin, C Kreidy, and Rona K. Graham
- Subjects
0301 basic medicine ,Olfactory system ,Neurogenesis ,Olfaction ,Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Huntington's disease ,Olfactory Mucosa ,Piriform cortex ,Genetics ,medicine ,Animals ,Humans ,Molecular Biology ,Genetics (clinical) ,Cell Proliferation ,Neurons ,Cell Death ,Brain ,General Medicine ,medicine.disease ,Olfactory Bulb ,Olfactory bulb ,Smell ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Huntington Disease ,Atrophy ,Neuroscience ,Neural development ,Olfactory epithelium ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
Olfactory dysfunction and altered neurogenesis are observed in several neurodegenerative disorders including Huntington disease (HD). These deficits occur early and correlate with a decline in global cognitive performance, depression and structural abnormalities of the olfactory system including the olfactory epithelium, bulb and cortices. However, the role of olfactory system dysfunction in the pathogenesis of HD remains poorly understood and the mechanisms underlying this dysfunction are unknown. We show that deficits in odour identification, discrimination and memory occur in HD individuals. Assessment of the olfactory system in an HD murine model demonstrates structural abnormalities in the olfactory bulb (OB) and piriform cortex, the primary cortical recipient of OB projections. Furthermore, a decrease in piriform neuronal counts and altered expression levels of neuronal nuclei and tyrosine hydroxylase in the OB are observed in the YAC128 HD model. Similar to the human HD condition, olfactory dysfunction is an early phenotype in the YAC128 mice and concurrent with caspase activation in the murine HD OB. These data provide a link between the structural olfactory brain region atrophy and olfactory dysfunction in HD and suggest that cell proliferation and cell death pathways are compromised and may contribute to the olfactory deficits in HD.
- Published
- 2020
40. Laquinimod Treatment Improves Myelination Deficits at the Transcriptional and Ultrastructural Levels in the YAC128 Mouse Model of Huntington Disease
- Author
-
Marta Garcia-Miralles, Michael R. Hayden, Mahmoud A. Pouladi, Liang Juin Tan, Jing Ying Tan, Carola I. Radulescu, Neta Zach, Haim Belinson, Harwin Sidik, and Nur Amirah Binte Mohammad Yusof
- Subjects
Male ,0301 basic medicine ,Neurology ,Transcription, Genetic ,Cell Count ,Striatum ,Quinolones ,Corpus callosum ,Corpus Callosum ,chemistry.chemical_compound ,0302 clinical medicine ,Myelin Sheath ,Behavior, Animal ,Depression ,Oligodendroglia ,Huntington Disease ,Phenotype ,medicine.anatomical_structure ,Antidepressant ,Female ,Microglia ,medicine.medical_specialty ,Neuroscience (miscellaneous) ,Mice, Transgenic ,Motor Activity ,White matter ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Atrophy ,Immune system ,Internal medicine ,Cytochrome P-450 CYP1A1 ,medicine ,Animals ,Humans ,Learning ,Inflammation ,business.industry ,medicine.disease ,Corpus Striatum ,Disease Models, Animal ,030104 developmental biology ,Endocrinology ,Gene Expression Regulation ,Receptors, Aryl Hydrocarbon ,chemistry ,Astrocytes ,business ,Laquinimod ,030217 neurology & neurosurgery - Abstract
Laquinimod, an immunomodulatory agent under clinical development for Huntington disease (HD), has recently been shown to confer behavioural improvements that are coupled with prevention of atrophy of the white matter (WM)-rich corpus callosum (CC) in the YAC128 HD mice. However, the nature of the WM improvements is not known yet. Here we investigated the effects of laquinimod on HD-related myelination deficits at the cellular, molecular and ultrastructural levels. We showed that laquinimod treatment improves motor learning and motor function deficits in YAC128 HD mice, and confirmed its antidepressant effect even at the lowest dose used. In addition, we demonstrated for the first time the beneficial effects of laquinimod on myelination in the posterior region of the CC where it reversed changes in myelin sheath thickness and rescued Mbp mRNA and protein deficits. Furthermore, the effect of laquinimod on myelin-related gene expression was not region-specific since the levels of the Mbp and Plp1 transcripts were also increased in the striatum. Also, we did not detect changes in immune cell densities or levels of inflammatory genes in 3-month-old YAC128 HD mice, and these were not altered with laquinimod treatment. Thus, the beneficial effects of laquinimod on HD-related myelination abnormalities in YAC128 HD mice do not appear to be dependent on its immunomodulatory activity. Altogether, our findings describe the beneficial effects of laquinimod treatment on HD-related myelination abnormalities and highlight its therapeutic potential for the treatment of WM pathology in HD patients.
- Published
- 2018
41. A thiol probe for measuring unfolded protein load and proteostasis in cells
- Author
-
Mahmoud A. Pouladi, Ben Zhong Tang, Danny M. Hatters, Leann Tilley, Zhegang Song, Rebecca J. Wood, Nagaraj S. Moily, Gavin E. Reid, Moore Z. Chen, Mona Radwan, Jessica L. Bridgford, Trevor A. Smith, Yuning Hong, and Xiaohong Xu
- Subjects
0301 basic medicine ,Protein Folding ,Huntingtin ,Proteome ,General Physics and Astronomy ,01 natural sciences ,Maleimides ,chemistry.chemical_compound ,Mice ,lcsh:Science ,chemistry.chemical_classification ,Huntingtin Protein ,Multidisciplinary ,Tunicamycin ,Artemisinins ,3. Good health ,Biochemistry ,Protein folding ,Oligopeptides ,Globular protein ,Cells ,Science ,Biology ,010402 general chemistry ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Animals ,Humans ,Parasites ,Cysteine ,Sulfhydryl Compounds ,Fluorescent Dyes ,General Chemistry ,0104 chemical sciences ,Malaria ,030104 developmental biology ,Proteostasis ,HEK293 Cells ,chemistry ,Solubility ,Molecular Probes ,Biophysics ,Mutant Proteins ,lcsh:Q ,HeLa Cells - Abstract
When proteostasis becomes unbalanced, unfolded proteins can accumulate and aggregate. Here we report that the dye, tetraphenylethene maleimide (TPE-MI) can be used to measure cellular unfolded protein load. TPE-MI fluorescence is activated upon labelling free cysteine thiols, normally buried in the core of globular proteins that are exposed upon unfolding. Crucially TPE-MI does not become fluorescent when conjugated to soluble glutathione. We find that TPE-MI fluorescence is enhanced upon reaction with cellular proteomes under conditions promoting accumulation of unfolded proteins. TPE-MI reactivity can be used to track which proteins expose more cysteine residues under stress through proteomic analysis. We show that TPE-MI can report imbalances in proteostasis in induced pluripotent stem cell models of Huntington disease, as well as cells transfected with mutant Huntington exon 1 before the formation of visible aggregates. TPE-MI also detects protein damage following dihydroartemisinin treatment of the malaria parasites Plasmodium falciparum. TPE-MI therefore holds promise as a tool to probe proteostasis mechanisms in disease., Proteostasis is maintained through a number of molecular mechanisms, some of which function to protect the folded state of proteins. Here the authors demonstrate the use of TPE-MI in a fluorigenic dye assay for the quantitation of unfolded proteins that can be used to assess proteostasis on a cellular or proteome scale.
- Published
- 2017
42. Reversal of Phenotypic Abnormalities by CRISPR/Cas9-Mediated Gene Correction in Huntington Disease Patient-Derived Induced Pluripotent Stem Cells
- Author
-
Florent Ginhoux, Xiaohong Xu, Yihui Huang, Mahmoud A. Pouladi, Jolene Ooi, Marie Loh, Donovan Low, Byrappa Venkatesh, Amin Ziaei, Yilin Tay, Carola I. Radulescu, Su-In Yoon, Bernice Sim, George J. Augustine, Bryan Tsong-Jye Ng, Kagistia Hana Utami, Alvin Yu Jin Ng, and Lee Kong Chian School of Medicine (LKCMedicine)
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Biochemistry ,hiPSC ,transcriptional dysrgulation ,disease modeling ,Gene expression ,genetic editing ,CRISPR ,Cell Self Renewal ,CAS9 ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,TRANSGENIC MICE ,Gene Editing ,Neurons ,Genetics ,lcsh:R5-920 ,human induced pluripotent stem cell (hiPSC) ,Gene Expression Regulation, Developmental ,Cell Differentiation ,DEFECTS ,Huntington disease ,Phenotype ,Mitochondria ,Cell biology ,DNA-Binding Proteins ,DIFFERENTIATION ,neurodegenerative disorders ,Gene Targeting ,lcsh:Medicine (General) ,Life Sciences & Biomedicine ,CHCHD2 ,MODELS ,Induced Pluripotent Stem Cells ,INHIBITION ,Biology ,Article ,Cell Line ,Mitochondrial Proteins ,03 medical and health sciences ,Cell & Tissue Engineering ,mitochondrial dysfunction ,medicine ,Humans ,Gene ,STRIATAL PROJECTION NEURONS ,Science & Technology ,MUTATIONS ,Growth factor ,Cell Biology ,Electrophysiological Phenomena ,030104 developmental biology ,lcsh:Biology (General) ,Forebrain ,CRISPR-Cas Systems ,disease phenotypes ,Trinucleotide repeat expansion ,Transcription Factors ,Developmental Biology - Abstract
Summary Huntington disease (HD) is a dominant neurodegenerative disorder caused by a CAG repeat expansion in HTT. Here we report correction of HD human induced pluripotent stem cells (hiPSCs) using a CRISPR-Cas9 and piggyBac transposon-based approach. We show that both HD and corrected isogenic hiPSCs can be differentiated into excitable, synaptically active forebrain neurons. We further demonstrate that phenotypic abnormalities in HD hiPSC-derived neural cells, including impaired neural rosette formation, increased susceptibility to growth factor withdrawal, and deficits in mitochondrial respiration, are rescued in isogenic controls. Importantly, using genome-wide expression analysis, we show that a number of apparent gene expression differences detected between HD and non-related healthy control lines are absent between HD and corrected lines, suggesting that these differences are likely related to genetic background rather than HD-specific effects. Our study demonstrates correction of HD hiPSCs and associated phenotypic abnormalities, and the importance of isogenic controls for disease modeling using hiPSCs., Graphical Abstract, Highlights • A CRISPR-Cas9 and PiggyBac-based approach allows efficient correction of HD hiPSCs • The corrected hiPSCs can be differentiated into synaptically active neurons • Correction of HD gene mutation reverses a number of phenotypic abnormalities • Isogenic hiPSCs help distinguish mutation from genetic background-related effects, Pouladi and colleagues show that genetic correction of HD human induced pluripotent stem cells using a CRISPR-Cas9 and piggyBac transposon-based approach rescues a number of phenotypic abnormalities in derived neural cells, including increased susceptibility to growth factor withdrawal and deficits in mitochondrial respiration, and helps distinguish HD-specific from genetic background-related molecular and cellular phenotypes.
- Published
- 2017
43. Expanded huntingtin CAG repeats disrupt the balance between neural progenitor expansion and differentiation in human cerebral organoids
- Author
-
Juergen A. Knoblich, Dong Shin Park, Florent Ginhoux, Shiming Ma, Sarah R. Langley, Kagistia Hana Utami, Chit Fang Cheok, Magdalena Renner, Obed Akwasi Aning, Jinqiu Zhang, Enrico Petretto, Mahmoud A. Pouladi, and Jolene Ooi
- Subjects
Neuroepithelial cell ,Huntingtin ,Neurogenesis ,Cell cycle ,Biology ,Progenitor cell ,Induced pluripotent stem cell ,Embryonic stem cell ,Progenitor ,Cell biology - Abstract
Huntington disease (HD) manifests in both adult and juvenile forms. Mutant HTT gene carriers are thought to undergo normal brain development followed by a degenerative phase, resulting in progressive clinical manifestations. However, recent studies in children and prodromal individuals at risk for HD have raised the possibility of abnormal neurodevelopment. Although key findings in rodent models support this notion, direct evidence in the context of human physiology remains lacking. Using a panel of isogenic HD human embryonic pluripotent stem cells and cerebral organoids, we investigated the impact of mutant HTT on early neurodevelopment. We find that ventricular zone-like neuroepithelial progenitor layer expansion is blunted in an HTT CAG repeat length-dependent manner due to premature neurogenesis in HD cerebral organoids, driven by cell intrinsic processes. Transcriptional profiling and imaging analysis revealed impaired cell cycle regulatory processes, increased G1 length, and increased asymmetric division of apical progenitors, collectively contributing to premature neuronal differentiation. We demonstrate increased activity of the ATM-p53 pathway, an up-stream regulator of cell cycle processes, and show that treatment with ATM antagonists partially rescues the blunted neuroepithelial progenitor expansion in HD organoids. Our findings suggest that CAG repeat length regulates the balance between neural progenitor expansion and differentiation during early neurodevelopment. Our results further support the view that HD, at least in its early-onset forms, may not be a purely neurodegenerative disorder, and that abnormal neurodevelopment may be a component of HD pathophysiology.
- Published
- 2019
44. Gene expression profiles complement the analysis of genomic modifiers of the clinical onset of Huntington disease
- Author
-
Michael R. Hayden, Nicholas S. Caron, Bernard Ng, Colin J. D. Ross, Lorenzo Casal, Mahmoud A. Pouladi, Sara Mostafavi, Galen E.B. Wright, Jolene Ooi, and Xiaohong Xu
- Subjects
Genetics ,0303 health sciences ,Disease ,Biology ,Genome ,3. Good health ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,Age of onset ,Cognitive decline ,Trinucleotide repeat expansion ,Gene ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Huntington disease (HD) is a neurodegenerative disorder that is caused by a CAG repeat expansion in the HTT gene. In an attempt to identify genomic modifiers that contribute towards the age of onset of HD, we performed a transcriptome wide association study assessing heritable differences in genetically determined expression in diverse tissues, employing genome wide data from over 4,000 patients. This identified genes that showed evidence for colocalization and replication, with downstream functional validation being performed in isogenic HD stem cells and patient brains. Enrichment analyses detected associations with various biologically-relevant gene sets and striatal coexpression modules that are mediated by CAG length. Further, cortical coexpression modules that are relevant for HD onset were also associated with cognitive decline and HD-related traits in a longitudinal cohort. In summary, the combination of population-scale gene expression information with HD patient genomic data identified novel modifier genes for the disorder.
- Published
- 2019
45. Intrinsic mutant HTT-mediated defects in oligodendroglia cause myelination deficits and behavioral abnormalities in Huntington disease
- Author
-
Marta Garcia-Miralles, Lisa M. Anderson, Ana Maria Rondelli, Nirmala Arul Rayan, Mahmoud A. Pouladi, Han-Gyu Bae, Michael R. Hayden, Nicholas S. Caron, Costanza Ferrari Bardile, Roy Tang Yi Teo, Anna Williams, Sangyong Jung, Shyam Prabhakar, Nathan Harmston, Sabine Waltl, Sarah R. Langley, Enrico Petretto, and Lee Kong Chian School of Medicine (LKCMedicine)
- Subjects
Huntingtin ,Population ,Disease ,White matter ,Mice ,Myelin ,medicine ,Animals ,Medicine [Science] ,Epigenetics ,education ,Pathological ,Huntingtin Protein ,education.field_of_study ,Multidisciplinary ,Behavior, Animal ,Microglia ,business.industry ,Polycomb Repressive Complex 2 ,Biological Sciences ,White Matter ,Mice, Mutant Strains ,Disease Models, Animal ,Oligodendroglia ,Huntington Disease ,medicine.anatomical_structure ,Mutation ,business ,Neuroscience ,Demyelinating Diseases - Abstract
White matter abnormalities are a nearly universal pathological feature of neurodegenerative disorders including Huntington disease (HD). A long-held assumption is that this white matter pathology is simply a secondary outcome of the progressive neuronal loss that manifests with advancing disease. Using a mouse model of HD, here we show that white matter and myelination abnormalities are an early disease feature appearing before the manifestation of any behavioral abnormalities or neuronal loss. We further show that selective inactivation of mutant huntingtin (mHTT) in the NG2+ oligodendrocyte progenitor cell population prevented myelin abnormalities and certain behavioral deficits in HD mice. Strikingly, the improvements in behavioral outcomes were seen despite the continued expression of mHTT in nonoligodendroglial cells including neurons, astrocytes, and microglia. Using RNA-seq and ChIP-seq analyses, we implicate a pathogenic mechanism that involves enhancement of polycomb repressive complex 2 (PRC2) activity by mHTT in the intrinsic oligodendroglial dysfunction and myelination deficits observed in HD. Our findings challenge the long-held dogma regarding the etiology of white matter pathology in HD and highlight the contribution of epigenetic mechanisms to the observed intrinsic oligodendroglial dysfunction. Our results further suggest that ameliorating white matter pathology and oligodendroglial dysfunction may be beneficial for HD. Agency for Science, Technology and Research (A*STAR) Accepted version We thank members of the M.A.P. laboratory for helpful discussions and comments. C.F.B. is supported by a Singapore International Graduate Award from the Agency for Science, Technology and Research (A*STAR). M.A.P. is supported by grants from A*STAR and the National University of Singapore.
- Published
- 2019
46. Huntingtin confers fitness but is not embryonically essential in zebrafish development
- Author
-
Mahmoud A. Pouladi, Christy J. Ang, and Harwin Sidik
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,Embryo, Nonmammalian ,Huntingtin ,Morpholino ,animal diseases ,Nerve Tissue Proteins ,Body size ,Biology ,Morpholinos ,Gene Knockout Techniques ,03 medical and health sciences ,0302 clinical medicine ,mental disorders ,Animals ,Body Size ,Humans ,Amino Acid Sequence ,Neurulation ,Molecular Biology ,Zebrafish ,Conserved Sequence ,Genetic Association Studies ,Gene knockout ,030304 developmental biology ,Gene Editing ,Huntingtin Protein ,0303 health sciences ,Sequence Homology, Amino Acid ,Wild type ,Cell Biology ,Zebrafish Proteins ,biology.organism_classification ,Embryonic stem cell ,Cell biology ,nervous system diseases ,nervous system ,Models, Animal ,Genetic Fitness ,CRISPR-Cas Systems ,Sequence Alignment ,030217 neurology & neurosurgery ,Function (biology) ,Developmental Biology - Abstract
Attempts to constitutively knockout HTT in rodents resulted in embryonic lethality, curtailing efforts to study HTT function later in development. Here we show that HTT is dispensable for early zebrafish development, contrasting published zebrafish morpholino experiment results. Homozygous HTT knockouts were embryonically viable and appeared developmentally normal through juvenile stages. Comparison of adult fish revealed significant reduction in body size and fitness in knockouts compared to hemizygotes and wildtype fish, indicating an important role for wildtype HTT in postnatal development. Our zebrafish model provides an opportunity to examine the function of wildtype HTT later in development.
- Published
- 2019
- Full Text
- View/download PDF
47. Impaired Remyelination in a Mouse Model of Huntington Disease
- Author
-
Mahmoud A. Pouladi, Yi Lin Tay, Roy Tang Yi Teo, Liang Juin Tan, Charbel A. Kreidy, Nur Amirah Binte Mohammad Yusof, and Costanza Ferrari Bardile
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Neurology ,Neuroscience (miscellaneous) ,Mice, Transgenic ,Disease ,Corpus callosum ,Corpus Callosum ,White matter ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Cuprizone ,0302 clinical medicine ,Internal medicine ,medicine ,Animals ,Humans ,Remyelination ,business.industry ,Oligodendrocyte ,Axons ,Staining ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Huntington Disease ,business ,Neuroglia ,030217 neurology & neurosurgery ,Demyelinating Diseases - Abstract
White matter (WM) abnormalities are a well-established feature of Huntington disease (HD), although their nature is not fully understood. Here, we asked whether remyelination as a measure of WM plasticity is impaired in a model of HD. Using the cuprizone assay, we examined demyelination and remyelination responses in YAC128 HD mice. Treatment with 0.2% cuprizone (CPZ) for 6 weeks resulted in significant reduction in mature (GSTπ-positive) oligodendrocyte counts and FluoroMyelin staining in the corpus callosum, leading to similar demyelination states in YAC128 and wild-type (WT) mice. Six weeks following cessation of CPZ, we observed robust remyelination in WT mice as indicated by an increase in mature oligodendrocyte counts and FluoroMyelin staining. In contrast, YAC128 mice exhibited an impaired remyelination response. The increase in mature oligodendrocyte counts in YAC128 HD mice following CPZ cessation was lower than that of WT. Furthermore, there was no increase in FluoroMyelin staining compared to the demyelinated state in YAC128 mice. We confirmed these findings using electron microscopy where the CPZ-induced reduction in myelinated axons was reversed following CPZ cessation in WT but not YAC128 mice. Our findings demonstrate that remyelination is impaired in YAC128 mice and suggest that WM plasticity may be compromised in HD.
- Published
- 2019
48. Novel mutation in HTRA1 in a family with diffuse white matter lesions and inflammatory features
- Author
-
Amin Ziaei, Xiaohong Xu, Leila Dehghani, Mahmoud A. Pouladi, Christof Haffner, Vahid Shaygannejad, Alvin Yu Jin Ng, Andreas Zellner, Bruno Reversade, Sumanty Tohari, Carine Bonnard, Byrappa Venkatesh, Center for Reproductive Medicine, ACS - Diabetes & metabolism, and Amsterdam Reproduction & Development
- Subjects
0301 basic medicine ,Mutation ,Pathology ,medicine.medical_specialty ,Ataxia ,business.industry ,Myelitis ,medicine.disease_cause ,Disease gene identification ,medicine.disease ,Hyperintensity ,Leukoencephalopathy ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Hair loss ,Germline mutation ,medicine ,Neurology (clinical) ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Genetics (clinical) - Abstract
ObjectiveTo investigate the possible involvement of germline mutations in a neurologic condition involving diffuse white matter lesions.MethodsThe patients were 3 siblings born to healthy parents. We performed homozygosity mapping, whole-exome sequencing, site-directed mutagenesis, and immunoblotting.ResultsAll 3 patients showed clinical manifestations of ataxia, behavioral and mood changes, premature hair loss, memory loss, and lower back pain. In addition, they presented with inflammatory-like features and recurrent rhinitis. MRI showed abnormal diffuse demyelination lesions in the brain and myelitis in the spinal cord. We identified an insertion in high-temperature requirement A (HTRA1), which showed complete segregation in the pedigree. Functional analysis showed the mutation to affect stability and secretion of truncated protein.ConclusionsThe patients' clinical manifestations are consistent with cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL; OMIM #600142), which is known to be caused by HTRA1 mutations. Because some aspects of the clinical presentation deviate from those reported for CARASIL, our study expands the spectrum of clinical consequences of loss-of-function mutations in HTRA1.
- Published
- 2019
49. Generation of four H1 hESC sublines carrying a hemizygous knock-out/mutant MECP2
- Author
-
Mahmoud A. Pouladi, Kim S. Robinson, Harwin Sidik, Ruizhu Zeng, Franklin L. Zhong, Bruno Reversade, Center for Reproductive Medicine, ACS - Diabetes & metabolism, Amsterdam Reproduction & Development (AR&D), ACS - Heart failure & arrhythmias, Reversade, Bruno, Zeng, Ruizhu, Sidik, Harwin, Robinson, Kim S., Zhong, Franklin L., Pouladiahi, Mahmoud A., School of Medicine, and Department of Medical Genetics
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Methyl-CpG-Binding Protein 2 ,Mutant ,Karyotype ,Rett syndrome ,Methyl-CpG-binding protein 2 ,Mutation ,MECP2 ,Cell Line ,03 medical and health sciences ,Exon ,0302 clinical medicine ,Neurodevelopmental disorder ,mental disorders ,medicine ,Rett Syndrome ,CRISPR ,Humans ,lcsh:QH301-705.5 ,Embryonic Stem Cells ,Genetics ,Gene Editing ,Nuclease ,biology ,Cas9 ,Cell Differentiation ,Cell Biology ,General Medicine ,Exons ,medicine.disease ,nervous system diseases ,030104 developmental biology ,lcsh:Biology (General) ,biology.protein ,Medicine ,CRISPR-Cas Systems ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Rett syndrome (RTT) is a childhood neurodevelopmental disorder caused by mutations in MECP2. To study the molecular mechanisms underlying RTT, four sublines of H1 hESCs were generated, carrying a hemizygous knockout or mutant allele of MECP2. Exons 3 and 4 of MECP2 were targeted using the CRISPR/Cas9 nuclease system., Strategic Positioning Fund for Genetic Orphan Diseases; SUREKids from the Agency for Science Technology and Research (Singapore); TLGM-NDD from Agency for Science Technology and Research (Singapore); the National University of Singapore
- Published
- 2019
50. Neurodevelopmental Defects: A Micropatterned Human‐Specific Neuroepithelial Tissue for Modeling Gene and Drug‐Induced Neurodevelopmental Defects (Adv. Sci. 5/2021)
- Author
-
Geetika Sahni, Kagistia Hana Utami, Puck Wee Chan, Thong Teck Tan, Jean Jacques Clement Fatien, Umut Altunoglu, Yi-Chin Toh, Carine Bonnard, Jeremy Teo Choon Meng, Jerome Zu Yao Tan, Bruno Reversade, Hülya Kayserili, Shu-Yung Chang, and Mahmoud A. Pouladi
- Subjects
Back Cover ,Drug ,General Chemical Engineering ,media_common.quotation_subject ,General Engineering ,Neuroepithelial Tissue ,General Physics and Astronomy ,Medicine (miscellaneous) ,Biology ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Cell biology ,General Materials Science ,Human specific ,Gene ,media_common - Abstract
In article number 2001100, Yi‐Chin Toh and co‐workers combine hPSC micropatterning and a temporally‐sequenced induction protocol to specify neuroepithelial (NE) cells in spatial juxtaposition to mesoendoderm cells. This results in the formation of a reproducible and highly‐organized NE tissue. By evaluating NE tissue structural dysmorphia in the micropatterned NE model, gene‐ and drug‐induced neurodevelopmental defects can be successfully modelled. [Image: see text]
- Published
- 2021
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.