1
|
Jiang H, Xiao Z, Saleem K, Zhong P, Li L, Chhetri G, Li P, Jiang Z, Yan Z, Feng J. Generation of human induced pluripotent stem cell-derived cortical neurons expressing the six tau isoforms. J Alzheimers Dis 2025:13872877251334831. [PMID: 40267294 DOI: 10.1177/13872877251334831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
BackgroundThe alternative splicing (AS) of MAPT, which encodes Tau, in the adult human brain produces six major isoforms that play critical roles in the pathogenesis of tauopathies including Alzheimer's disease. Previous efforts have failed to differentiate human induced pluripotent stem cells (hiPSCs) to cortical neurons expressing the six isoforms of Tau.ObjectiveWe aim to develop a differentiation method capable of producing the six Tau isoforms in hiPSC-derived cortical neurons.MethodsWe searched for the optimal concentration, duration and treatment window of morphogens in the differentiation of hiPSCs through embryoid bodies (EBs) to dorsal forebrain neuroepithelial cells then to cortical neurons.ResultsThe combined inhibition of WNT, SHH, and SMAD signaling in EBs generated neuroepithelial cells expressing appropriate dorsal forebrain markers, while suppressing ventral, midbrain, and hindbrain genes. Further differentiation in neurogenic and neurotrophic factors produced MAP2+ neurons at day 18. The iPSC-derived neurons expressed markers of all cortical layers and exhibited synapse formation and synaptic physiology. In addition, MAP2+ neurons and mitotic cells expressing radial glial markers formed aggregates that could be dissociated to produce mature neurons with similar properties. Most importantly, the six Tau isoforms were expressed from day 80 in a developmentally regulated manner, modeling the situation in human brains on an accelerated timeline.ConclusionsThis chemically defined differentiation method produces a key hallmark of mature human cortical neurons by expressing the six main splicing isoforms of Tau. It will greatly facilitate disease modeling and therapeutic discovery for many human brain disorders involving cortical neurons.
Collapse
Affiliation(s)
- Houbo Jiang
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Zichun Xiao
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Komal Saleem
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Ping Zhong
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Li Li
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Gaurav Chhetri
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Pei Li
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Zhongjiao Jiang
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| | - Jian Feng
- Department of Physiology and Biophysics, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
2
|
Sozen B, Tam PPL, Pera MF. Pluripotent cell states and fates in human embryo models. Development 2025; 152:dev204565. [PMID: 40171916 PMCID: PMC11993252 DOI: 10.1242/dev.204565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
Pluripotency, the capacity to generate all cells of the body, is a defining property of a transient population of epiblast cells found in pre-, peri- and post-implantation mammalian embryos. As development progresses, the epiblast cells undergo dynamic transitions in pluripotency states, concurrent with the specification of extra-embryonic and embryonic lineages. Recently, stem cell-based models of pre- and post-implantation human embryonic development have been developed using stem cells that capture key properties of the epiblast at different developmental stages. Here, we review early primate development, comparing pluripotency states of the epiblast in vivo with cultured pluripotent cells representative of these states. We consider how the pluripotency status of the starting cells influences the development of human embryo models and, in turn, what we can learn about the human pluripotent epiblast. Finally, we discuss the limitations of these models and questions arising from the pioneering studies in this emerging field.
Collapse
Affiliation(s)
- Berna Sozen
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT 06501, USA
- Yale Stem Cell Center, Yale University, New Haven, CT 06520, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, Yale University, New Haven, CT 06501, USA
| | - Patrick P. L. Tam
- Embryology Research Unit, Children's Medical Research Institute and School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Martin F. Pera
- The Jackson Laboratory, Mammalian Genetics, Bar Harbor, ME 04609, USA
| |
Collapse
|
3
|
Artegiani B, Hendriks D. Organoids from pluripotent stem cells and human tissues: When two cultures meet each other. Dev Cell 2025; 60:493-511. [PMID: 39999776 DOI: 10.1016/j.devcel.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/13/2024] [Accepted: 01/10/2025] [Indexed: 02/27/2025]
Abstract
Human organoids are a widely used tool in cell biology to study homeostatic processes, disease, and development. The term organoids covers a plethora of model systems from different cellular origins that each have unique features and applications but bring their own challenges. This review discusses the basic principles underlying organoids generated from pluripotent stem cells (PSCs) as well as those derived from tissue stem cells (TSCs). We consider how well PSC- and TSC-organoids mimic the different intended organs in terms of cellular complexity, maturity, functionality, and the ongoing efforts to constitute predictive complex models of in vivo situations. We discuss the advantages and limitations associated with each system to answer different biological questions including in the field of cancer and developmental biology, and with respect to implementing emerging advanced technologies, such as (spatial) -omics analyses, CRISPR screens, and high-content imaging screens. We postulate how the two fields may move forward together, integrating advantages of one to the other.
Collapse
Affiliation(s)
| | - Delilah Hendriks
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| |
Collapse
|
4
|
Glass MR, Matoba N, Beltran AA, Patel NK, Farah TM, Eswar K, Bhargava S, Huang K, Curtin I, Ahmed S, Srivastava M, Drake E, Davis LT, Yeturi M, Sun K, Love MI, Simon JM, St John T, Marrus N, Pandey J, Estes A, Dager S, Schultz RT, Botteron K, Evans A, Kim SH, Styner M, McKinstry RC, Collins DL, Volk H, Benke K, Zwaigenbaum L, Hazlett H, Beltran AS, Girault JB, Shen MD, Piven J, Stein JL. Early cell cycle genes in cortical organoid progenitors predict interindividual variability in infant brain growth trajectories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637106. [PMID: 39974968 PMCID: PMC11839139 DOI: 10.1101/2025.02.07.637106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Human induced pluripotent stem cell (iPSC) derived cortical organoids (hCOs) model neurogenesis on an individual's genetic background. The degree to which hCO phenotypes recapitulate the brain growth of the participants from which they were derived is not well established. We generated up to 3 iPSC clones from each of 18 participants in the Infant Brain Imaging Study, who have undergone longitudinal brain imaging during infancy. We identified consistent hCO morphology and cortical cell types across clones from the same participant. hCO cross-sectional area and production of cortical hem cells were associated with in vivo cortical growth rates. Cell cycle associated genes expression in early progenitors at the crux of fate decision trajectories were correlated with cortical growth rate from 6-12 months of age, and were enriched in microcephaly and neurodevelopmental disorder genes. Our data suggest the hCOs capture inter-individual variation in cortical cell types influencing infant cortical surface area expansion.
Collapse
Affiliation(s)
- Madison R Glass
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors contributed equally
| | - Nana Matoba
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors contributed equally
| | - Alvaro A Beltran
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Niyanta K Patel
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Tala M Farah
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Karthik Eswar
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Shivam Bhargava
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Karen Huang
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Ian Curtin
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Sara Ahmed
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Mary Srivastava
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Emma Drake
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Liam T Davis
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Meghana Yeturi
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Kexin Sun
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Michael I Love
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Jeremy M Simon
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Present address: Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Present address: Department of Biostatistics, Harvard T.H. Chan School of Public Health, MA, USA
| | - Tanya St John
- Department of Speech and Hearing Sciences, University of Washington; Seattle, WA, USA
- University of Washington Autism Center, University of Washington; Seattle, WA, USA
| | - Natasha Marrus
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO, USA
| | - Juhi Pandey
- Department of Psychiatry, University of Pennsylvania; Philadelphia, PA, USA
- Center for Autism Research, Children's Hospital of Philadelphia; Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania; Philadelphia, USA
| | - Annette Estes
- Department of Speech and Hearing Sciences, University of Washington; Seattle, WA, USA
- University of Washington Autism Center, University of Washington; Seattle, WA, USA
| | - Stephen Dager
- Department of Radiology, University of Washington; Seattle, WA, USA
- Department of Bioengineering, University of Washington; Seattle, WA, USA
| | - Robert T Schultz
- Department of Psychiatry, University of Pennsylvania; Philadelphia, PA, USA
- Center for Autism Research, Children's Hospital of Philadelphia; Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania; Philadelphia, USA
| | - Kelly Botteron
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO, USA
| | - Alan Evans
- Department of Neurology and Neurosurgery, McGill University; Montreal, QC, CA
- Department of Psychiatry, McGill University; Montreal, QC, CA
- Department of Biomedical Engineering, McGill University; Montreal, QC, CA
| | - Sun Hyung Kim
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Martin Styner
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Computer Sciences, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Robert C McKinstry
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO, USA
| | - D Louis Collins
- Department of Neurology and Neurosurgery, McGill University; Montreal, QC, CA
- Department of Biomedical Engineering, McGill University; Montreal, QC, CA
| | - Heather Volk
- School of Public Health, Johns Hopkins; Baltimore, MD, USA
| | - Kelly Benke
- School of Public Health, Johns Hopkins; Baltimore, MD, USA
| | - Lonnie Zwaigenbaum
- Department of Developmental Pediatrics, University of Alberta; Edmonton, AB, CA
| | - Heather Hazlett
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Adriana S Beltran
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Jessica B Girault
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors jointly supervised
| | - Mark D Shen
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors jointly supervised
| | - Joseph Piven
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors jointly supervised
| | - Jason L Stein
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors jointly supervised
- Lead contact
| |
Collapse
|
5
|
Fang K, Pishva E, Piers T, Scholpp S. Amyloid-β can activate JNK signalling via WNT5A-ROR2 to reduce synapse formation in Alzheimer's disease. J Cell Sci 2025; 138:JCS263526. [PMID: 39907042 PMCID: PMC11832185 DOI: 10.1242/jcs.263526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/19/2024] [Indexed: 02/06/2025] Open
Abstract
Wnt signalling is an essential signalling system in neurogenesis, with a crucial role in synaptic plasticity and neuronal survival, processes that are disrupted in Alzheimer's disease (AD). Within this network, the Wnt/β-catenin pathway has been studied for its neuroprotective role, and this is suppressed in AD. However, the involvement of the non-canonical Wnt-planar cell polarity (Wnt/PCP) pathway in AD remains to be determined. This study investigates the role of ROR2, a Wnt/PCP co-receptor, in synaptogenesis. We demonstrate that WNT5A-ROR2 signalling activates the JNK pathway, leading to synapse loss in mature neurons. This effect mirrors the synaptotoxic actions of Aβ1-42 and DKK1, which are elevated in AD. Notably, blocking ROR2 and JNK mitigates Aβ1-42 and DKK1-induced synapse loss, suggesting their dependence on ROR2. In induced pluripotent stem cell (iPSC)-derived cortical neurons carrying a PSEN1 mutation, known to increase the Aβ42/40 ratio, we observed increased WNT5A-ROR2 clustering and reduced numbers of synapses. Inhibiting ROR2 or JNK partially rescued synaptogenesis in these neurons. These findings suggest that, unlike the Wnt/β-catenin pathway, the Wnt/PCP-ROR2 signalling pathway can operate in a feedback loop with Aβ1-42 to enhance JNK signalling and contribute to synapse loss in AD.
Collapse
Affiliation(s)
- Kevin Fang
- Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| | - Ehsan Pishva
- Department of Psychiatry and Neuropsychology, Mental Health and Neuroscience Research Institute, University Maastricht, 6229 ER Maastricht, The Netherlands
| | - Thomas Piers
- Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
- University of Exeter Medical School, RILD Building, RD&E Hospital Wonford, Exeter EX2 5DW, UK
| | - Steffen Scholpp
- Living Systems Institute, University of Exeter, Exeter EX4 4QD, UK
| |
Collapse
|
6
|
De Beuckeleer S, Van De Looverbosch T, Van Den Daele J, Ponsaerts P, De Vos WH. Unbiased identification of cell identity in dense mixed neural cultures. eLife 2025; 13:RP95273. [PMID: 39819559 PMCID: PMC11741521 DOI: 10.7554/elife.95273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025] Open
Abstract
Induced pluripotent stem cell (iPSC) technology is revolutionizing cell biology. However, the variability between individual iPSC lines and the lack of efficient technology to comprehensively characterize iPSC-derived cell types hinder its adoption in routine preclinical screening settings. To facilitate the validation of iPSC-derived cell culture composition, we have implemented an imaging assay based on cell painting and convolutional neural networks to recognize cell types in dense and mixed cultures with high fidelity. We have benchmarked our approach using pure and mixed cultures of neuroblastoma and astrocytoma cell lines and attained a classification accuracy above 96%. Through iterative data erosion, we found that inputs containing the nuclear region of interest and its close environment, allow achieving equally high classification accuracy as inputs containing the whole cell for semi-confluent cultures and preserved prediction accuracy even in very dense cultures. We then applied this regionally restricted cell profiling approach to evaluate the differentiation status of iPSC-derived neural cultures, by determining the ratio of postmitotic neurons and neural progenitors. We found that the cell-based prediction significantly outperformed an approach in which the population-level time in culture was used as a classification criterion (96% vs 86%, respectively). In mixed iPSC-derived neuronal cultures, microglia could be unequivocally discriminated from neurons, regardless of their reactivity state, and a tiered strategy allowed for further distinguishing activated from non-activated cell states, albeit with lower accuracy. Thus, morphological single-cell profiling provides a means to quantify cell composition in complex mixed neural cultures and holds promise for use in the quality control of iPSC-derived cell culture models.
Collapse
Affiliation(s)
- Sarah De Beuckeleer
- Laboratory of Cell Biology and Histology, University of AntwerpAntwerpBelgium
| | | | | | - Peter Ponsaerts
- Laboratory of Experimental Haematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of AntwerpAntwerpBelgium
| | - Winnok H De Vos
- Laboratory of Cell Biology and Histology, University of AntwerpAntwerpBelgium
- Antwerp Centre for Advanced Microscopy, University of AntwerpAntwerpBelgium
- µNeuro Research Centre of Excellence, University of AntwerpAntwerpBelgium
| |
Collapse
|
7
|
Lai D, Sosicka P, Williams DJ, Bowyer ME, Ressler AK, Kohrt SE, Muron SJ, Crino PB, Freeze HH, Boland MJ, Heinzen EL. SLC35A2 loss of function variants affect glycomic signatures, neuronal fate, and network dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.27.630524. [PMID: 39763953 PMCID: PMC11703275 DOI: 10.1101/2024.12.27.630524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
SLC35A2 encodes a UDP-galactose transporter essential for glycosylation of proteins and galactosylation of lipids and glycosaminoglycans. Germline genetic SLC35A2 variants have been identified in congenital disorders of glycosylation and somatic SLC35A2 variants have been linked to intractable epilepsy associated with malformations of cortical development. However, the functional consequences of these pathogenic variants on brain development and network integrity remain elusive. In this study, we use an isogenic human induced pluripotent stem cell-derived neuron model to comprehensively interrogate the functional impact of loss of function variants in SLC35A2 through the integration of cellular and molecular biology, protein glycosylation analysis, neural network dynamics, and single cell electrophysiology. We show that loss of function variants in SLC35A2 result in disrupted glycomic signatures and precocious neurodevelopment, yielding hypoactive, asynchronous neural networks. This aberrant network activity is attributed to an inhibitory/excitatory imbalance as characterization of neural composition revealed preferential differentiation of SLC35A2 loss of function variants towards the GABAergic fate. Additionally, electrophysiological recordings of synaptic activity reveal a shift in excitatory/inhibitory balance towards increased inhibitory drive, indicating changes occurring specifically at the pre-synaptic terminal. Our study is the first to provide mechanistic insight regarding the early development and functional connectivity of SLC35A2 loss of function variant harboring human neurons, providing important groundwork for future exploration of potential therapeutic interventions.
Collapse
Affiliation(s)
- Dulcie Lai
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Paulina Sosicka
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Damian J Williams
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - MaryAnn E Bowyer
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Andrew K Ressler
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Sarah E Kohrt
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Savannah J Muron
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Peter B Crino
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Hudson H Freeze
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, 92037, USA
| | - Michael J Boland
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Center for Epilepsy and Neurodevelopmental Disorders, Perelman School of Medicine, University of Pennsylvania, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Erin L Heinzen
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, 27599, USA
- Institute for Genomic Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| |
Collapse
|
8
|
Naghshbandieh A, Naghshbandieh A, Barfi E, Abkhooie L. Assessment of the level of apoptosis in differentiated pseudo-neuronal cells derived from neural stem cells under the influence of various inducers. AMERICAN JOURNAL OF STEM CELLS 2024; 13:250-270. [PMID: 39850017 PMCID: PMC11751472 DOI: 10.62347/bptg6174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/23/2024] [Indexed: 01/25/2025]
Abstract
Development and maintenance of the nervous system are governed by a scheduled cell death mechanism known as apoptosis. Very much how neurons survive and function depends on the degree of death in differentiating pseudo-neuronal cells produced from neural stem cells. Different inducers can affect the degree of death in these cells: hormones, medicines, growth factors, and others. Developing inventive therapies for neurodegenerative illnesses depends on a knowledge of how these inducers impact mortality in differentiated pseudo-neuronal cells. Using flow cytometry, Western blotting, and fluorescence microscopy among other techniques, the degree of death in many pseudo-neuronal cells is evaluated. Flow cytometry generates dead cell counts from measurements of cell size, granularity, and DNA content. Whereas fluorescence microscopy visualizes dead cells using fluorescent dyes or antibodies, Western blotting detects caspases and Bcl-2 family proteins. This review attempts to offer a thorough investigation of present studies on death in differentiated pseudo-neuronal cells produced from neural stem cells under the effect of different inducers. Through investigating how these inducers influence death, the review aims to provide information that might direct the next studies and support treatment plans for neurodegenerative diseases. With an eye toward inducers like retinoic acid, selegiline, cytokines, valproic acid, and small compounds, we examined research to evaluate death rates. The findings offer important new perspectives on the molecular processes guiding death in these cells. There is still a complete lack of understanding of how different factors affect the molecular processes that lead to death, so understanding these processes can contribute to new therapeutic approaches to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Adele Naghshbandieh
- Department of Anatomical Sciences, School of Medical Sciences, Tarbiat Modares UniversityTehran, Iran
| | - Atefe Naghshbandieh
- Department of Pharmaceutical Biotechnology and Department of Pharmaceutical and Bimolecular Science, University of MilanMilan, Italy
| | - Elahe Barfi
- Razi Herbal Medicines Research Center, Lorestan University of Medical SciencesKhorramabad, Iran
| | - Leila Abkhooie
- Razi Herbal Medicines Research Center, Lorestan University of Medical SciencesKhorramabad, Iran
- Department of Medical Biotechnology, School of Medicine, Lorestan University of Medical SciencesKhorramabad, Iran
| |
Collapse
|
9
|
Bates SM, Evans KV, Delsing L, Wong R, Cornish G, Bahjat M. Immune safety challenges facing the preclinical assessment and clinical progression of cell therapies. Drug Discov Today 2024; 29:104239. [PMID: 39521331 DOI: 10.1016/j.drudis.2024.104239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 10/15/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
The promise of curative outcomes for life-limiting diseases using cell therapies is starting to become a reality, not only for patients with end-stage cancer, but also increasingly for regenerative therapies, including dentistry, ocular, neurodegenerative, and cardiac diseases. The introduction of often genetically modified cells into a patient can come with an extensive range of safety considerations. From an immune perspective, cell-based therapies carry inherent consequences and consideration of factors, such as the cell source (donor-derived autologous cells versus allogeneic cells), the intrinsic cellular nature of the therapy, and engineering/manufacturing methods, all of which influence the likelihood of inducing unwanted immune responses. Here, we provide an overview of the potential immune safety risks associated with cell therapies and explore possible mitigation approaches.
Collapse
Affiliation(s)
- Stephanie M Bates
- Safety Innovation, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Kelly V Evans
- Safety Innovation, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Louise Delsing
- Cell and Gene Therapy Safety, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Ryan Wong
- Cell and Gene Therapy Safety, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Georgina Cornish
- Oncology Safety, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Mahnoush Bahjat
- Safety Innovation, Clinical Pharmacology and Safety Sciences, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
10
|
Piers TM, Fang K, Namboori SC, Liddle C, Rogers S, Bhinge A, Killick R, Scholpp S. WNT7A-positive dendritic cytonemes control synaptogenesis in cortical neurons. Development 2024; 151:dev202868. [PMID: 39576204 DOI: 10.1242/dev.202868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 11/07/2024] [Indexed: 12/10/2024]
Abstract
Synaptogenesis involves the transformation of dendritic filopodial contacts into stable connections with the exact apposition of synaptic components. Signalling triggered by Wnt/β-catenin and calcium has been postulated to aid this process. However, it is unclear how such a signalling process orchestrates synapse formation to organise the spatial arrangement of synapses along dendrites. We show that WNT7A is loaded on dynamic dendritic filopodia during spine formation in human cortical neurons. WNT7A is present at the tips of the filopodia and the contact sites with dendrites of neighbouring neurons, triggering spatially restricted localisation of the Wnt co-receptor LRP6. Here, we demonstrate that WNT7A at filopodia tips leads to the induction of calcium transients, the clustering of pre- and postsynaptic proteins, and the subsequent transformation into mature spines. Although soluble WNT7A protein can also support synaptogenesis, it fails to provide this degree of spatial information for spine formation and calcium transients, and synaptic markers are induced ectopically along the dendrites. Our data suggest that dendritic filopodia are WNT7A-bearing cytonemes required for focal calcium signalling and initiation of synapse formation, and provide an elegant mechanism for orchestrating the positioning of synapses along dendrites.
Collapse
Affiliation(s)
- Thomas M Piers
- Living Systems Institute, Department of Biosciences, Faculty of Health and Life Sciences, University of Exeter, EX4 4QD, UK
| | - Kevin Fang
- Bioimaging Centre, Department of Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter EX4 4QD, UK
| | - Seema C Namboori
- Living Systems Institute, Department of Biosciences, Faculty of Health and Life Sciences, University of Exeter, EX4 4QD, UK
| | - Corin Liddle
- Bioimaging Centre, Department of Biosciences, Faculty of Health and Life Sciences, University of Exeter, Exeter EX4 4QD, UK
| | - Sally Rogers
- Living Systems Institute, Department of Biosciences, Faculty of Health and Life Sciences, University of Exeter, EX4 4QD, UK
| | - Akshay Bhinge
- Living Systems Institute, Department of Biosciences, Faculty of Health and Life Sciences, University of Exeter, EX4 4QD, UK
| | - Richard Killick
- Living Systems Institute, Department of Biosciences, Faculty of Health and Life Sciences, University of Exeter, EX4 4QD, UK
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London SE5 8AF, UK
| | - Steffen Scholpp
- Living Systems Institute, Department of Biosciences, Faculty of Health and Life Sciences, University of Exeter, EX4 4QD, UK
| |
Collapse
|
11
|
Whye D, Norabuena EM, Srinivasan GR, Wood D, Polanco TJ, Makhortova NR, Sahin M, Buttermore ED. A Hybrid 2D-to-3D in vitro Differentiation Platform Improves Outcomes of Cerebral Cortical Organoid Generation in hiPSCs. Curr Protoc 2024; 4:e70022. [PMID: 39400999 DOI: 10.1002/cpz1.70022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Three-dimensional (3D) cerebral cortical organoids are popular in vitro cellular model systems widely used to study human brain development and disease, compared to traditional stem cell-derived methods that use two-dimensional (2D) monolayer cultures. Despite the advancements made in protocol development for cerebral cortical organoid derivation over the past decade, limitations due to biological, mechanistic, and technical variables remain in generating these complex 3D cellular systems. Building from our previously established differentiation system, we have made modifications to our existing 3D cerebral cortical organoid protocol that resolve several of these technical and biological challenges when working with diverse groups of human induced pluripotent stem cell (hiPSC) lines. This improved protocol blends a 2D monolayer culture format for the specification of neural stem cells and expansion of neuroepithelial progenitor cells with a 3D system for improved self-aggregation and subsequent organoid development. Furthermore, this "hybrid" approach is amenable to both an accelerated cerebral cortical organoid protocol as well as an alternative long-term differentiation protocol. In addition to establishing a hybrid technical format, this protocol also offers phenotypic and morphological characterization of stage-specific cellular profiles using antibodies and fluorescent-based dyes for live cell imaging. © 2024 Wiley Periodicals LLC. Basic Protocol 1: hiPSC-based 2D monolayer specification into neural stem cells (NSCs) Basic Protocol 2: Serial passaging and 2D monolayer expansion of neuroepithelial progenitor cells (NPCs) Support Protocol 1: Direct cryopreservation and rapid thawing of NSCs and NPCs Basic Protocol 3: Bulk aggregation of 3D neurospheres and accelerated cerebral cortical organoid differentiation Alternate Protocol 1: Bulk aggregation of 3D neurospheres and long-term cerebral cortical organoid differentiation Support Protocol 2: High-throughput 3D neurosphere formation and 2D neurosphere migration assay Support Protocol 3: LIVE/DEAD stain cell imaging assay of 3D neurospheres Support Protocol 4: NeuroFluor NeuO live cell dye for 3D cerebral cortical organoids.
Collapse
Affiliation(s)
- Dosh Whye
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| | - Erika M Norabuena
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| | - Gayathri Rajaram Srinivasan
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| | - Delaney Wood
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| | - Taryn J Polanco
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| | - Nina R Makhortova
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
- Department of Neurology, Harvard Medical School, Boston, Massachusetts
| | - Mustafa Sahin
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
- Department of Neurology, Harvard Medical School, Boston, Massachusetts
| | - Elizabeth D Buttermore
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts
- F.M. Kirby Neurobiology Department, Boston Children's Hospital, Boston, Massachusetts
| |
Collapse
|
12
|
Wang X, Lalli M, Thopte U, Buxbaum JD. A scalable, high-throughput neural development platform identifies shared impact of ASD genes on cell fate and differentiation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614184. [PMID: 39386704 PMCID: PMC11463611 DOI: 10.1101/2024.09.25.614184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Background Deleterious mutations in hundreds of genes confer high risk for neurodevelopmental disorders (NDDs), posing significant challenges for therapeutic development. Identifying convergent pathways shared across NDD genes could reveal high-impact therapeutic targets. Methods To identity convergent pathways in NDD genes, we optimized Perturb-seq, a method combining CRISPR perturbation with single-cell RNA sequencing (scRNA-seq), and applied structural topic modeling (STM) to simultaneously assess impact on cell fate and developmental stage. We then studied a subset of autism spectrum disorder (ASD) genes implicated in regulation of gene expression using these improved molecular and analytical approaches. Results Results from targeting 60 high-confidence ASD risk genes revealed significant effects on neural development. As expected, ASD risk genes impacted both progenitor fate and/or neuronal differentiation. Using STM, we could identify latent topics jointly capturing cell types, cell fate, and differentiation stages. Repression of ASD risk genes led to changes in topic proportions and effects of four genes (DEAF1, KMT2A, MED13L, and MYT1L) were validated in an independent dataset. Conclusions Our optimized Perturb-seq method, combined with a novel analytical approach, provides a powerful, cost-effective framework for uncovering convergent mechanisms among genes involved in complex neurodevelopmental processes. Application of these methods advanced understanding of the impact of ASD mutations on multiple dimensions of neural development, and provides a framework for a broader examination of the function of NDD risk genes.
Collapse
Affiliation(s)
- Xuran Wang
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York NY, USA; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York NY, USA
| | - Matthew Lalli
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Urvashi Thopte
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph D. Buxbaum
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York NY, USA; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York NY, USA
| |
Collapse
|
13
|
Shan X, Zhang A, Rezzonico MG, Tsai MC, Sanchez-Priego C, Zhang Y, Chen MB, Choi M, Andrade López JM, Phu L, Cramer AL, Zhang Q, Pattison JM, Rose CM, Hoogenraad CC, Jeong CG. Fully defined NGN2 neuron protocol reveals diverse signatures of neuronal maturation. CELL REPORTS METHODS 2024; 4:100858. [PMID: 39255791 PMCID: PMC11440061 DOI: 10.1016/j.crmeth.2024.100858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 04/26/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024]
Abstract
NGN2-driven induced pluripotent stem cell (iPSC)-to-neuron conversion is a popular method for human neurological disease modeling. In this study, we present a standardized approach for generating neurons utilizing clonal, targeted-engineered iPSC lines with defined reagents. We demonstrate consistent production of excitatory neurons at scale and long-term maintenance for at least 150 days. Temporal omics, electrophysiological, and morphological profiling indicate continued maturation to postnatal-like neurons. Quantitative characterizations through transcriptomic, imaging, and functional assays reveal coordinated actions of multiple pathways that drive neuronal maturation. We also show the expression of disease-related genes in these neurons to demonstrate the relevance of our protocol for modeling neurological disorders. Finally, we demonstrate efficient generation of NGN2-integrated iPSC lines. These workflows, profiling data, and functional characterizations enable the development of reproducible human in vitro models of neurological disorders.
Collapse
Affiliation(s)
- Xiwei Shan
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Ai Zhang
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Mitchell G Rezzonico
- Department of OMNI Bioinformatics, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Ming-Chi Tsai
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | | | - Yingjie Zhang
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Michelle B Chen
- Department of Cellular and Tissue Genomics, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Meena Choi
- Department of Proteomic and Genomic Technologies, Genentech, Inc., South San Francisco, CA 94080, USA
| | | | - Lilian Phu
- Department of Proteomic and Genomic Technologies, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Amber L Cramer
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Qiao Zhang
- Department of Discovery Oncology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Jillian M Pattison
- Advanced Cell Engineering, Department of Molecular Biology, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Christopher M Rose
- Department of Proteomic and Genomic Technologies, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Casper C Hoogenraad
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Claire G Jeong
- Department of Neuroscience, Genentech, Inc., South San Francisco, CA 94080, USA.
| |
Collapse
|
14
|
Kim SK, Seo S, Stein-O'Brien G, Jaishankar A, Ogawa K, Micali N, Luria V, Karger A, Wang Y, Kim H, Hyde TM, Kleinman JE, Voss T, Fertig EJ, Shin JH, Bürli R, Cross AJ, Brandon NJ, Weinberger DR, Chenoweth JG, Hoeppner DJ, Sestan N, Colantuoni C, McKay RD. Individual variation in the emergence of anterior-to-posterior neural fates from human pluripotent stem cells. Stem Cell Reports 2024; 19:1336-1350. [PMID: 39151428 PMCID: PMC11411333 DOI: 10.1016/j.stemcr.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 08/19/2024] Open
Abstract
Variability between human pluripotent stem cell (hPSC) lines remains a challenge and opportunity in biomedicine. In this study, hPSC lines from multiple donors were differentiated toward neuroectoderm and mesendoderm lineages. We revealed dynamic transcriptomic patterns that delineate the emergence of these lineages, which were conserved across lines, along with individual line-specific transcriptional signatures that were invariant throughout differentiation. These transcriptomic signatures predicted an antagonism between SOX21-driven forebrain fates and retinoic acid-induced hindbrain fates. Replicate lines and paired adult tissue demonstrated the stability of these line-specific transcriptomic traits. We show that this transcriptomic variation in lineage bias had both genetic and epigenetic origins, aligned with the anterior-to-posterior structure of early mammalian development, and was present across a large collection of hPSC lines. These findings contribute to developing systematic analyses of PSCs to define the origin and consequences of variation in the early events orchestrating individual human development.
Collapse
Affiliation(s)
- Suel-Kee Kim
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Seungmae Seo
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | | | - Amritha Jaishankar
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Kazuya Ogawa
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Nicola Micali
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA; Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Victor Luria
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA; Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Amir Karger
- IT-Research Computing, Harvard Medical School, Boston, MA 02115, USA
| | - Yanhong Wang
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Hyojin Kim
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA; Departments of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Departments of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Joel E Kleinman
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA; Departments of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Ty Voss
- Division of Preclinical Innovation, Nation Center for Advancing Translational Science, NIH, Bethesda, MD 20892, USA
| | - Elana J Fertig
- Departments of Oncology, Biomedical Engineering, and Applied Mathematics and Statistics, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Joo-Heon Shin
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Roland Bürli
- Astra-Zeneca Neuroscience iMED., 141 Portland Street, Cambridge, MA 01239, USA
| | - Alan J Cross
- Astra-Zeneca Neuroscience iMED., 141 Portland Street, Cambridge, MA 01239, USA
| | - Nicholas J Brandon
- Astra-Zeneca Neuroscience iMED., 141 Portland Street, Cambridge, MA 01239, USA
| | - Daniel R Weinberger
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA; Departments of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Departments of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Departments of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Joshua G Chenoweth
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Daniel J Hoeppner
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA
| | - Nenad Sestan
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06510, USA; Departments of Genetics, Psychiatry, and Comparative Medicine, Kavli Institute for Neuroscience, Program in Cellular Neuroscience, Neurodegeneration and Repair, Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA.
| | - Carlo Colantuoni
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA; Departments of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Departments of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA; Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Ronald D McKay
- Lieber Institute for Brain Development, 855 North Wolfe Street, Baltimore, MD 21205, USA; Departments of Cell Biology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
15
|
Choi J, Gang S, Ramalingam M, Hwang J, Jeong H, Yoo J, Cho HH, Kim BC, Jang G, Jeong HS, Jang S. BML-281 promotes neuronal differentiation by modulating Wnt/Ca 2+ and Wnt/PCP signaling pathway. Mol Cell Biochem 2024; 479:2391-2403. [PMID: 37768498 DOI: 10.1007/s11010-023-04857-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023]
Abstract
Histone deacetylase (HDAC) inhibitors promote differentiation through post-translational modifications of histones. BML-281, an HDAC6 inhibitor, has been known to prevent tumors, acute dextran sodium sulfate-associated colitis, and lung injury. However, the neurogenic differentiation effect of BML-281 is poorly understood. In this study, we investigated the effect of BML-281 on neuroblastoma SH-SY5Y cell differentiation into mature neurons by immunocytochemistry (ICC), reverse transcriptase PCR (RT-PCR), quantitative PCR (qPCR), and western blotting analysis. We found that the cells treated with BML-281 showed neurite outgrowth and morphological changes into mature neurons under a microscope. It was confirmed that the gene expression of neuronal markers (NEFL, MAP2, Tuj1, NEFH, and NEFM) was increased with certain concentrations of BML-281. Similarly, the protein expression of neuronal markers (NeuN, Synaptophysin, Tuj1, and NFH) was upregulated with BML-281 compared to untreated cells. Following treatment with BML-281, the expression of Wnt5α increased, and downstream pathways were activated. Interestingly, both Wnt/Ca2+ and Wnt/PCP pathways activated and regulated PKC, Cdc42, RhoA, Rac1/2/3, and p-JNK. Therefore, BML-281 induces the differentiation of SH-SY5Y cells into mature neurons by activating the non-canonical Wnt signaling pathway. From these results, we concluded that BML-281 might be a novel drug to differentiation into neuronal cells through the regulation of Wnt signaling pathway to reduce the neuronal cell death.
Collapse
Affiliation(s)
- Jiyun Choi
- Department of Physiology, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea
| | - Seoyeon Gang
- Department of Physiology, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea
- Department of Pre-Medical Science, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea
| | - Mahesh Ramalingam
- Department of Physiology, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea
| | - Jinsu Hwang
- Department of Physiology, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea
| | - Haewon Jeong
- Department of Physiology, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea
| | - Jin Yoo
- Department of Physiological Education, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyong-Ho Cho
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Byeong C Kim
- Department of Neurology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Geupil Jang
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Han-Seong Jeong
- Department of Physiology, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea.
| | - Sujeong Jang
- Department of Physiology, Chonnam National University Medical School, Jellanamdo, 58128, Republic of Korea.
| |
Collapse
|
16
|
Schuster J, Lu X, Dang Y, Klar J, Wenz A, Dahl N, Chen X. Epigenetic insights into GABAergic development in Dravet Syndrome iPSC and therapeutic implications. eLife 2024; 12:RP92599. [PMID: 39190448 PMCID: PMC11349296 DOI: 10.7554/elife.92599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024] Open
Abstract
Dravet syndrome (DS) is a devastating early-onset refractory epilepsy syndrome caused by variants in the SCN1A gene. A disturbed GABAergic interneuron function is implicated in the progression to DS but the underlying developmental and pathophysiological mechanisms remain elusive, in particularly at the chromatin level. Induced pluripotent stem cells (iPSCs) derived from DS cases and healthy donors were used to model disease-associated epigenetic abnormalities of GABAergic development. Chromatin accessibility was assessed at multiple time points (Day 0, Day 19, Day 35, and Day 65) of GABAergic differentiation. Additionally, the effects of the commonly used anti-seizure drug valproic acid (VPA) on chromatin accessibility were elucidated in GABAergic cells. The distinct dynamics in the chromatin profile of DS iPSC predicted accelerated early GABAergic development, evident at D19, and diverged further from the pattern in control iPSC with continued differentiation, indicating a disrupted GABAergic maturation. Exposure to VPA at D65 reshaped the chromatin landscape at a variable extent in different iPSC-lines and rescued the observed dysfunctional development of some DS iPSC-GABA. The comprehensive investigation on the chromatin landscape of GABAergic differentiation in DS-patient iPSC offers valuable insights into the epigenetic dysregulations associated with interneuronal dysfunction in DS. Moreover, the detailed analysis of the chromatin changes induced by VPA in iPSC-GABA holds the potential to improve the development of personalized and targeted anti-epileptic therapies.
Collapse
Affiliation(s)
- Jens Schuster
- Department of Immunology, Genetics and Pathology, Uppsala University and Science for Life LaboratoryUppsalaSweden
| | - Xi Lu
- Department of Immunology, Genetics and Pathology, Uppsala University and Science for Life LaboratoryUppsalaSweden
| | - Yonglong Dang
- Department of Immunology, Genetics and Pathology, Uppsala University and Science for Life LaboratoryUppsalaSweden
| | - Joakim Klar
- Department of Immunology, Genetics and Pathology, Uppsala University and Science for Life LaboratoryUppsalaSweden
| | - Amelie Wenz
- Department of Immunology, Genetics and Pathology, Uppsala University and Science for Life LaboratoryUppsalaSweden
| | - Niklas Dahl
- Department of Immunology, Genetics and Pathology, Uppsala University and Science for Life LaboratoryUppsalaSweden
| | - Xingqi Chen
- Department of Immunology, Genetics and Pathology, Uppsala University and Science for Life LaboratoryUppsalaSweden
| |
Collapse
|
17
|
St Clair-Glover M, Finol-Urdaneta RK, Maddock M, Wallace E, Miellet S, Wallace G, Yue Z, Dottori M. Efficient fabrication of 3D bioprinted functional sensory neurons using an inducible Neurogenin-2 human pluripotent stem cell line. Biofabrication 2024; 16:045022. [PMID: 39084624 DOI: 10.1088/1758-5090/ad69c4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 07/31/2024] [Indexed: 08/02/2024]
Abstract
Three-dimensional (3D) tissue models have gained recognition for their improved ability to mimic the native cell microenvironment compared to traditional two-dimensional models. This progress has been driven by advances in tissue-engineering technologies such as 3D bioprinting, a promising method for fabricating biomimetic living tissues. While bioprinting has succeeded in generating various tissues to date, creating neural tissue models remains challenging. In this context, we present an accelerated approach to fabricate 3D sensory neuron (SN) structures using a transgenic human pluripotent stem cell (hPSC)-line that contains an inducible Neurogenin-2 (NGN2) expression cassette. The NGN2 hPSC line was first differentiated to neural crest cell (NCC) progenitors, then incorporated into a cytocompatible gelatin methacryloyl-based bioink for 3D bioprinting. Upregulated NGN2 expression in the bioprinted NCCs resulted in induced SN (iSN) populations that exhibited specific cell markers, with 3D analysis revealing widespread neurite outgrowth through the scaffold volume. Calcium imaging demonstrated functional activity of iSNs, including membrane excitability properties and voltage-gated sodium channel (NaV) activity. This efficient approach to generate 3D bioprinted iSN structures streamlines the development of neural tissue models, useful for the study of neurodevelopment and disease states and offering translational potential.
Collapse
Affiliation(s)
- Mitchell St Clair-Glover
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, NSW 2522, Australia
| | - Rocio K Finol-Urdaneta
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Marnie Maddock
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Eileen Wallace
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, NSW 2522, Australia
| | - Sara Miellet
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Gordon Wallace
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, NSW 2522, Australia
| | - Zhilian Yue
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, NSW 2522, Australia
| | - Mirella Dottori
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong, NSW 2522, Australia
- School of Medical, Indigenous, and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, University of Wollongong, NSW 2522, Australia
| |
Collapse
|
18
|
Hung C, Fertan E, Livesey FJ, Klenerman D, Patani R. APP antisense oligonucleotides reduce amyloid-β aggregation and rescue endolysosomal dysfunction in Alzheimer's disease. Brain 2024; 147:2325-2333. [PMID: 38527856 PMCID: PMC11224613 DOI: 10.1093/brain/awae092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/23/2024] [Accepted: 02/29/2024] [Indexed: 03/27/2024] Open
Abstract
APP gene dosage is strongly associated with Alzheimer's disease (AD) pathogenesis. Genomic duplication of the APP locus leads to autosomal dominant early-onset AD. Individuals with Down syndrome (trisomy of chromosome 21) harbour three copies of the APP gene and invariably develop progressive AD with highly characteristic neuropathological features. Restoring expression of APP to the equivalent of that of two gene copies, or lower, is a rational therapeutic strategy, as it would restore physiological levels of neuronal APP protein without the potentially deleterious consequences of inadvertently inducing loss of APP function. Here we find that antisense oligonucleotides (ASOs) targeting APP are an effective approach to reduce APP protein levels and rescue endolysosome and autophagy dysfunction in APP duplication and Trisomy 21 human induced pluripotent stem cell (hiPSC)-derived cortical neurons. Importantly, using ultrasensitive single-aggregate imaging techniques, we show that APP targeting ASOs significantly reduce both intracellular and extracellular amyloid-β-containing aggregates. Our results highlight the potential of APP ASOs as a therapeutic approach for forms of AD caused by duplication of the APP gene, including monogenic AD and AD related to Down syndrome.
Collapse
Affiliation(s)
- Christy Hung
- Human Stem Cells and Neurodegeneration Laboratory, The Francis Crick Institute, London NW1 1AT, UK
- Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, London WC1N 1DZ, UK
| | - Emre Fertan
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Frederick J Livesey
- Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, London WC1N 1DZ, UK
| | - David Klenerman
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- UK Dementia Research Institute at University of Cambridge, Cambridge CB2 0XY, UK
| | - Rickie Patani
- Human Stem Cells and Neurodegeneration Laboratory, The Francis Crick Institute, London NW1 1AT, UK
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| |
Collapse
|
19
|
Glenn RA, Do SC, Guruvayurappan K, Corrigan EK, Santini L, Medina-Cano D, Singer S, Cho H, Liu J, Broman K, Czechanski A, Reinholdt L, Koche R, Furuta Y, Kunz M, Vierbuchen T. A PLURIPOTENT STEM CELL PLATFORM FOR IN VITRO SYSTEMS GENETICS STUDIES OF MOUSE DEVELOPMENT. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.06.597758. [PMID: 38895226 PMCID: PMC11185710 DOI: 10.1101/2024.06.06.597758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The directed differentiation of pluripotent stem cells (PSCs) from panels of genetically diverse individuals is emerging as a powerful experimental system for characterizing the impact of natural genetic variation on developing cell types and tissues. Here, we establish new PSC lines and experimental approaches for modeling embryonic development in a genetically diverse, outbred mouse stock (Diversity Outbred mice). We show that a range of inbred and outbred PSC lines can be stably maintained in the primed pluripotent state (epiblast stem cells -- EpiSCs) and establish the contribution of genetic variation to phenotypic differences in gene regulation and directed differentiation. Using pooled in vitro fertilization, we generate and characterize a genetic reference panel of Diversity Outbred PSCs (n = 230). Finally, we demonstrate the feasibility of pooled culture of Diversity Outbred EpiSCs as "cell villages", which can facilitate the differentiation of large numbers of EpiSC lines for forward genetic screens. These data can complement and inform similar efforts within the stem cell biology and human genetics communities to model the impact of natural genetic variation on phenotypic variation and disease-risk.
Collapse
Affiliation(s)
- Rachel A. Glenn
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Cell and Developmental Biology Program, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY, USA
| | - Stephanie C. Do
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Emily K. Corrigan
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Present address: Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA and Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Laura Santini
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel Medina-Cano
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sarah Singer
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hyein Cho
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jing Liu
- Mouse Genetics Core Facility, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karl Broman
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI USA
| | | | | | - Richard Koche
- Center for Epigenetics Research, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yasuhide Furuta
- Mouse Genetics Core Facility, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Meik Kunz
- The Bioinformatics CRO, Sanford Florida, 32771 USA
| | - Thomas Vierbuchen
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Center for Stem Cell Biology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
20
|
Zhao H, Sun R, Wu L, Huang P, Liu W, Ma Q, Liao Q, Du J. Bioinformatics Identification and Experimental Validation of a Prognostic Model for the Survival of Lung Squamous Cell Carcinoma Patients. Biochem Genet 2024:10.1007/s10528-024-10828-z. [PMID: 38806973 DOI: 10.1007/s10528-024-10828-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 05/08/2024] [Indexed: 05/30/2024]
Abstract
Lung squamous cell carcinoma (LUSC) kills more than four million people yearly. Creating more trustworthy tumor molecular markers for LUSC early detection, diagnosis, prognosis, and customized treatment is essential. Cuproptosis, a novel form of cell death, opened up a new field of study for searching for trustworthy tumor indicators. Our goal was to build a risk model to assess drug sensitivity, monitor immune function, and predict prognosis in LUSC patients. The 19 cuproptosis-related genes were found in the literature, and patient genomic and clinical information was collected using the Cancer Genomic Atlas (TCGA) database. The LUSC patients were grouped using unsupervised clustering techniques, and 7626 differentially expressed genes were identified. Using univariate COX analysis, LASSO regression analysis, and multivariate COX analysis, a prognostic model for LUSC patients was developed. The tumor immune escape was evaluated using the Tumor Immune Dysfunction and Exclusion (TIDE) method. The R packages 'pRRophetic,' 'ggpubr,' and 'ggplot2' were utilized to examine drug sensitivity. For modeling, a 6-cuproptosis-based gene signature was found. Patients with high-risk LUSC had significantly worse survival rates than those with low-risk conditions. The possibility of tumor immunological escape was increased in patients with higher risk scores due to more immune cell inactivation. For patients with high-risk LUSC, we discovered seven potent potential drugs (AZD6482, CHIR.99021, CMK, Embelin, FTI.277, Imatinib, and Pazopanib). In conclusion, the cuproptosis-based genes predictive risk model can be utilized to predict outcomes, track immune function, and evaluate medication sensitivity in LUSC patients.
Collapse
Affiliation(s)
- Hongtao Zhao
- Department of Immunology, College of Basic Medicine, Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Ruonan Sun
- Department of Immunology, College of Basic Medicine, Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Lei Wu
- College of Department of Information and Library Science, Guilin Medical University, Guilin, 541004, China
| | - Peiluo Huang
- Department of Immunology, College of Basic Medicine, Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Wenjing Liu
- Department of Immunology, College of Basic Medicine, Guilin Medical University, Guilin, 541199, Guangxi, China
| | - Qiuhong Ma
- Department of Clinical Laboratory, Zibo Central Hospital, Zibo, 255036, China.
| | - Qinyuan Liao
- Department of Immunology, College of Basic Medicine, Guilin Medical University, Guilin, 541199, Guangxi, China.
| | - Juan Du
- Department of Immunology, College of Basic Medicine, Guilin Medical University, Guilin, 541199, Guangxi, China.
| |
Collapse
|
21
|
Vo QD, Saito Y, Ida T, Nakamura K, Yuasa S. The use of artificial intelligence in induced pluripotent stem cell-based technology over 10-year period: A systematic scoping review. PLoS One 2024; 19:e0302537. [PMID: 38771829 PMCID: PMC11108174 DOI: 10.1371/journal.pone.0302537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/09/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Stem cell research, particularly in the domain of induced pluripotent stem cell (iPSC) technology, has shown significant progress. The integration of artificial intelligence (AI), especially machine learning (ML) and deep learning (DL), has played a pivotal role in refining iPSC classification, monitoring cell functionality, and conducting genetic analysis. These enhancements are broadening the applications of iPSC technology in disease modelling, drug screening, and regenerative medicine. This review aims to explore the role of AI in the advancement of iPSC research. METHODS In December 2023, data were collected from three electronic databases (PubMed, Web of Science, and Science Direct) to investigate the application of AI technology in iPSC processing. RESULTS This systematic scoping review encompassed 79 studies that met the inclusion criteria. The number of research studies in this area has increased over time, with the United States emerging as a leading contributor in this field. AI technologies have been diversely applied in iPSC technology, encompassing the classification of cell types, assessment of disease-specific phenotypes in iPSC-derived cells, and the facilitation of drug screening using iPSC. The precision of AI methodologies has improved significantly in recent years, creating a foundation for future advancements in iPSC-based technologies. CONCLUSIONS Our review offers insights into the role of AI in regenerative and personalized medicine, highlighting both challenges and opportunities. Although still in its early stages, AI technologies show significant promise in advancing our understanding of disease progression and development, paving the way for future clinical applications.
Collapse
Affiliation(s)
- Quan Duy Vo
- Faculty of Medicine, Department of Cardiovascular Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
- Faculty of Medicine, Nguyen Tat Thanh University, Ho Chi Minh City, Viet Nam
| | - Yukihiro Saito
- Department of Cardiovascular Medicine, Okayama University Hospital, Okayama, Japan
| | - Toshihiro Ida
- Faculty of Medicine, Department of Cardiovascular Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Kazufumi Nakamura
- Faculty of Medicine, Department of Cardiovascular Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Shinsuke Yuasa
- Faculty of Medicine, Department of Cardiovascular Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
22
|
Pottmeier P, Nikolantonaki D, Lanner F, Peuckert C, Jazin E. Sex-biased gene expression during neural differentiation of human embryonic stem cells. Front Cell Dev Biol 2024; 12:1341373. [PMID: 38764741 PMCID: PMC11101176 DOI: 10.3389/fcell.2024.1341373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 04/16/2024] [Indexed: 05/21/2024] Open
Abstract
Sex differences in the developing human brain are primarily attributed to hormonal influence. Recently however, genetic differences and their impact on the developing nervous system have attracted increased attention. To understand genetically driven sexual dimorphisms in neurodevelopment, we investigated genome-wide gene expression in an in vitro differentiation model of male and female human embryonic stem cell lines (hESC), independent of the effects of human sex hormones. Four male and four female-derived hESC lines were differentiated into a population of mixed neurons over 37 days. Differential gene expression and gene set enrichment analyses were conducted on bulk RNA sequencing data. While similar differentiation tendencies in all cell lines demonstrated the robustness and reproducibility of our differentiation protocol, we found sex-biased gene expression already in undifferentiated ESCs at day 0, but most profoundly after 37 days of differentiation. Male and female cell lines exhibited sex-biased expression of genes involved in neurodevelopment, suggesting that sex influences the differentiation trajectory. Interestingly, the highest contribution to sex differences was found to arise from the male transcriptome, involving both Y chromosome and autosomal genes. We propose 13 sex-biased candidate genes (10 upregulated in male cell lines and 3 in female lines) that are likely to affect neuronal development. Additionally, we confirmed gene dosage compensation of X/Y homologs escaping X chromosome inactivation through their Y homologs and identified a significant overexpression of the Y-linked demethylase UTY and KDM5D in male hESC during neuron development, confirming previous results in neural stem cells. Our results suggest that genetic sex differences affect neuronal differentiation trajectories, which could ultimately contribute to sex biases during human brain development.
Collapse
Affiliation(s)
- Philipp Pottmeier
- Department of Organismal Biology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
| | - Danai Nikolantonaki
- Department of Organismal Biology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
| | - Fredrik Lanner
- Division of Obstetrics and Gynecology, Department of Clinical Science, Intervention and Technology, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Christiane Peuckert
- Department of Organismal Biology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Elena Jazin
- Department of Organismal Biology, Evolutionary Biology Centre, Uppsala University, Uppsala, Sweden
| |
Collapse
|
23
|
Kuroda T, Yasuda S, Matsuyama S, Miura T, Sawada R, Matsuyama A, Yamamoto Y, Morioka MS, Kawaji H, Kasukawa T, Itoh M, Akutsu H, Kawai J, Sato Y. ROR2 expression predicts human induced pluripotent stem cell differentiation into neural stem/progenitor cells and GABAergic neurons. Sci Rep 2024; 14:690. [PMID: 38184695 PMCID: PMC10771438 DOI: 10.1038/s41598-023-51082-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 12/30/2023] [Indexed: 01/08/2024] Open
Abstract
Despite the development of various in vitro differentiation protocols for the efficient derivation of specific cell types, human induced pluripotent stem cell (hiPSC) lines have varing ability to differentiate into specific lineages. Therefore, surrogate markers for accurately predicting the differentiation propensity of hiPSC lines may facilitate cell-based therapeutic product development and manufacture. We attempted to identify marker genes that could predict the differentiation propensity of hiPSCs into neural stem/progenitor cells (NS/PCs). Using Spearman's rank correlation coefficients, we investigated genes in the undifferentiated state, the expression levels of which were significantly correlated with the neuronal differentiation propensity of several hiPSC lines. Among genes significantly correlated with NS/PC differentiation (P < 0.01), we identified ROR2 as a novel predictive marker. ROR2 expression in hiPSCs was negatively correlated with NS/PC differentiation tendency, regardless of the differentiation method, whereas its knockdown enhanced differentiation. ROR2 regulates NS/PC differentiation, suggesting that ROR2 is functionally essential for NS/PC differentiation. Selecting cell lines with relatively low ROR2 expression facilitated identification of hiPSCs that can differentiate into NS/PCs. Cells with ROR2 knockdown showed increased efficiency of differentiation into forebrain GABAergic neurons compared to controls. These findings suggest that ROR2 is a surrogate marker for selecting hiPSC lines appropriate for NS/PC and GABAergic neuronal differentiations.
Collapse
Affiliation(s)
- Takuya Kuroda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
- Life Science Technology Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Kanagawa, Japan
| | - Satoshi Yasuda
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
- Life Science Technology Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Kanagawa, Japan
- Department of Quality Assurance Science for Pharmaceuticals, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Satoko Matsuyama
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
- Center for Reverse TR, Osaka Habikino Medical Center, Osaka Prefectural Hospital Organization, Habikino, Osaka, Japan
| | - Takumi Miura
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
- Life Science Technology Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Kanagawa, Japan
- Center for Regenerative Medicine, National Center for Child Health and Development, Setagaya, Tokyo, Japan
| | - Rumi Sawada
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan
| | - Akifumi Matsuyama
- Center for Reverse TR, Osaka Habikino Medical Center, Osaka Prefectural Hospital Organization, Habikino, Osaka, Japan
| | - Yumiko Yamamoto
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | | | - Hideya Kawaji
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Research Center for Genome and Medical Sciences, Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo, Japan
| | - Takeya Kasukawa
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Masayoshi Itoh
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Hidenori Akutsu
- Center for Regenerative Medicine, National Center for Child Health and Development, Setagaya, Tokyo, Japan
| | - Jun Kawai
- Life Science Technology Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Kanagawa, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, Kawasaki, Kanagawa, Japan.
- Life Science Technology Project, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Kanagawa, Japan.
- Department of Quality Assurance Science for Pharmaceuticals, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan.
- Division of Drugs, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-9501, Japan.
- Department of Cellular and Gene Therapy Products, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
24
|
St George-Hyslop F, Haneklaus M, Kivisild T, Livesey FJ. Loss of CNTNAP2 Alters Human Cortical Excitatory Neuron Differentiation and Neural Network Development. Biol Psychiatry 2023; 94:780-791. [PMID: 37001843 DOI: 10.1016/j.biopsych.2023.03.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 05/14/2023]
Abstract
BACKGROUND Loss-of-function mutations in the contactin-associated protein-like 2 (CNTNAP2) gene are causal for neurodevelopmental disorders, including autism, schizophrenia, epilepsy, and intellectual disability. CNTNAP2 encodes CASPR2, a single-pass transmembrane protein that belongs to the neurexin family of cell adhesion molecules. These proteins have a variety of functions in developing neurons, including connecting presynaptic and postsynaptic neurons, and mediating signaling across the synapse. METHODS To study the effect of loss of CNTNAP2 function on human cerebral cortex development, and how this contributes to the pathogenesis of neurodevelopmental disorders, we generated human induced pluripotent stem cells from one neurotypical control donor null for full-length CNTNAP2, modeling cortical development from neurogenesis through to neural network formation in vitro. RESULTS CNTNAP2 is particularly highly expressed in the first two populations of early-born excitatory cortical neurons, and loss of CNTNAP2 shifted the relative proportions of these two neuronal types. Live imaging of excitatory neuronal growth showed that loss of CNTNAP2 reduced neurite branching and overall neuronal complexity. At the network level, developing cortical excitatory networks null for CNTNAP2 had complex changes in activity compared with isogenic controls: an initial period of relatively reduced activity compared with isogenic controls, followed by a lengthy period of hyperexcitability, and then a further switch to reduced activity. CONCLUSIONS Complete loss of CNTNAP2 contributes to the pathogenesis of neurodevelopmental disorders through complex changes in several aspects of human cerebral cortex excitatory neuron development that culminate in aberrant neural network formation and function.
Collapse
Affiliation(s)
- Frances St George-Hyslop
- University College London Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, University College London, London, United Kingdom
| | - Moritz Haneklaus
- University College London Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, University College London, London, United Kingdom
| | - Toomas Kivisild
- Estonian Biocentre, Institute of Genomics, University of Tartu, Tartu, Estonia; Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Frederick J Livesey
- University College London Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, University College London, London, United Kingdom.
| |
Collapse
|
25
|
Cavaleiro C, Afonso GJM, Oliveira PJ, Valero J, Mota SI, Ferreiro E. Urine-derived stem cells in neurological diseases: current state-of-the-art and future directions. Front Mol Neurosci 2023; 16:1229728. [PMID: 37965041 PMCID: PMC10642248 DOI: 10.3389/fnmol.2023.1229728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 10/10/2023] [Indexed: 11/16/2023] Open
Abstract
Stem cells have potential applications in the field of neurological diseases, as they allow for the development of new biological models. These models can improve our understanding of the underlying pathologies and facilitate the screening of new therapeutics in the context of precision medicine. Stem cells have also been applied in clinical tests to repair tissues and improve functional recovery. Nevertheless, although promising, commonly used stem cells display some limitations that curb the scope of their applications, such as the difficulty of obtention. In that regard, urine-derived cells can be reprogrammed into induced pluripotent stem cells (iPSCs). However, their obtaining can be challenging due to the low yield and complexity of the multi-phased and typically expensive differentiation protocols. As an alternative, urine-derived stem cells (UDSCs), included within the population of urine-derived cells, present a mesenchymal-like phenotype and have shown promising properties for similar purposes. Importantly, UDSCs have been differentiated into neuronal-like cells, auspicious for disease modeling, while overcoming some of the shortcomings presented by other stem cells for these purposes. Thus, this review assesses the current state and future perspectives regarding the potential of UDSCs in the ambit of neurological diseases, both for disease modeling and therapeutic applications.
Collapse
Affiliation(s)
- Carla Cavaleiro
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), Coimbra, Portugal
| | - Gonçalo J. M. Afonso
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), Coimbra, Portugal
| | - Paulo J. Oliveira
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Jorge Valero
- Instituto de Neurociencias de Castilla y León, University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Department of Cell Biology and Pathology, University of Salamanca, Salamanca, Spain
| | - Sandra I. Mota
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Elisabete Ferreiro
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
26
|
Simorgh S, Mousavi SA, To SK, Pasque V, Wierda K, Vervliet T, Yeganeh M, Pooyan P, Chai YC, Verfaillie C, Baharvand H. A facile method to generate cerebral organoids from human pluripotent stem cells. EXCLI JOURNAL 2023; 22:1055-1076. [PMID: 37927348 PMCID: PMC10620858 DOI: 10.17179/excli2023-6299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023]
Abstract
Human cerebral organoids (COs) are self-organizing three-dimensional (3D) neural structures that provide a human-specific platform to study the cellular and molecular processes that underlie different neurological events. The first step of CO generation from human pluripotent stem cells (hPSCs) is neural induction, which is an in vitro simulation of neural ectoderm development. Several signaling pathways cooperate during neural ectoderm development and in vitro differentiation of hPSCs toward neural cell lineages is also affected by them. In this study, we considered some of the known sources of these variable signaling cues arising from cell culture media components and sought to modulate their effects by applying a comprehensive combination of small molecules and growth factors for CO generation. Histological analysis demonstrated that these COs recapitulate the neural progenitor zone and early cortical layer organization, containing different types of neuronal and glial cells which was in accordance with single-nucleus transcriptome profiling results. Moreover, patch clamp and intracellular Ca2+ dynamic studies demonstrated that the COs behave as a functional neural network. Thus, this method serves as a facile protocol for generating hPSC-derived COs that faithfully mimic the features of their in vivo counterparts in the developing human brain. See also Figure 1(Fig. 1).
Collapse
Affiliation(s)
- Susan Simorgh
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven 3000, Belgium
| | - Seyed Ahmad Mousavi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - San Kit To
- Department of Development and Regeneration, Lab for Epigenetic Reprogramming, Leuven Stem Cell Institute, Leuven Single-Cell Omics Institute and Leuven Cancer Institute, KU Leuven-University of Leuven, Leuven 3000, Belgium
| | - Vincent Pasque
- Department of Development and Regeneration, Lab for Epigenetic Reprogramming, Leuven Stem Cell Institute, Leuven Single-Cell Omics Institute and Leuven Cancer Institute, KU Leuven-University of Leuven, Leuven 3000, Belgium
| | - Keimpe Wierda
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium
- Electrophysiology Unit, Leuven 3000, Belgium
| | - Tim Vervliet
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, KU Leuven, Leuven 3000, Belgium
| | - Meghdad Yeganeh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Paria Pooyan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Yoke Chin Chai
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven 3000, Belgium
| | - Catherine Verfaillie
- Stem Cell Institute, Department of Development and Regeneration, KU Leuven, Leuven 3000, Belgium
| | - Hossein Baharvand
- Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
27
|
Pollen AA, Kilik U, Lowe CB, Camp JG. Human-specific genetics: new tools to explore the molecular and cellular basis of human evolution. Nat Rev Genet 2023; 24:687-711. [PMID: 36737647 PMCID: PMC9897628 DOI: 10.1038/s41576-022-00568-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2022] [Indexed: 02/05/2023]
Abstract
Our ancestors acquired morphological, cognitive and metabolic modifications that enabled humans to colonize diverse habitats, develop extraordinary technologies and reshape the biosphere. Understanding the genetic, developmental and molecular bases for these changes will provide insights into how we became human. Connecting human-specific genetic changes to species differences has been challenging owing to an abundance of low-effect size genetic changes, limited descriptions of phenotypic differences across development at the level of cell types and lack of experimental models. Emerging approaches for single-cell sequencing, genetic manipulation and stem cell culture now support descriptive and functional studies in defined cell types with a human or ape genetic background. In this Review, we describe how the sequencing of genomes from modern and archaic hominins, great apes and other primates is revealing human-specific genetic changes and how new molecular and cellular approaches - including cell atlases and organoids - are enabling exploration of the candidate causal factors that underlie human-specific traits.
Collapse
Affiliation(s)
- Alex A Pollen
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| | - Umut Kilik
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Craig B Lowe
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.
| | - J Gray Camp
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland.
- University of Basel, Basel, Switzerland.
| |
Collapse
|
28
|
Tam RW, Keung AJ. Profiling transcriptomic responses of human stem cell-derived medium spiny neuron-like cells to exogenous phasic and tonic neurotransmitters. Mol Cell Neurosci 2023; 126:103876. [PMID: 37385515 PMCID: PMC10528483 DOI: 10.1016/j.mcn.2023.103876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 06/06/2023] [Accepted: 06/24/2023] [Indexed: 07/01/2023] Open
Abstract
Transcriptomic responses to neurotransmitters contribute to the complex processes driving memory and addiction. Advances in both measurement methods and experimental models continue to improve our understanding of this regulatory layer. Here we focus on the experimental potential of stem cell derived neurons, currently the only ethical model that can be used in reductionist and experimentally perturbable studies of human cells. Prior work has focused on generating distinct cell types from human stem cells, and has also shown their utility in modeling development and cellular phenotypes related to neurodegeneration. Here we seek an understanding of how stem cell derived neural cultures respond to perturbations experienced during development and disease progression. This work profiles transcriptomic responses of human medium spiny neuron-like cells with three specific goals. We first characterize transcriptomic responses to dopamine and dopamine receptor agonists and antagonists presented in dosing patterns mimicking acute, chronic, and withdrawal regimens. We also assess transcriptomic responses to low and persistent tonic levels of dopamine, acetylcholine, and glutamate to better mimic the in vivo environment. Finally, we identify similar and distinct responses between hMSN-like cells derived from H9 and H1 stem cell lines, providing some context for the extent of variability these types of systems will likely pose for experimentalists. The results here suggest future optimizations of human stem cell derived neurons to increase their in vivo relevance and the biological insights that can be garnered from these models.
Collapse
Affiliation(s)
- Ryan W Tam
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27606, United States of America
| | - Albert J Keung
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27606, United States of America.
| |
Collapse
|
29
|
Bertucci T, Bowles KR, Lotz S, Qi L, Stevens K, Goderie SK, Borden S, Oja LM, Lane K, Lotz R, Lotz H, Chowdhury R, Joy S, Arduini BL, Butler DC, Miller M, Baron H, Sandhof CA, Silva MC, Haggarty SJ, Karch CM, Geschwind DH, Goate AM, Temple S. Improved Protocol for Reproducible Human Cortical Organoids Reveals Early Alterations in Metabolism with MAPT Mutations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.11.548571. [PMID: 37503195 PMCID: PMC10369860 DOI: 10.1101/2023.07.11.548571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Cerebral cortical-enriched organoids derived from human pluripotent stem cells (hPSCs) are valuable models for studying neurodevelopment, disease mechanisms, and therapeutic development. However, recognized limitations include the high variability of organoids across hPSC donor lines and experimental replicates. We report a 96-slitwell method for efficient, scalable, reproducible cortical organoid production. When hPSCs were cultured with controlled-release FGF2 and an SB431542 concentration appropriate for their TGFBR1 / ALK5 expression level, organoid cortical patterning and reproducibility were significantly improved. Well-patterned organoids included 16 neuronal and glial subtypes by single cell RNA sequencing (scRNA-seq), frequent neural progenitor rosettes and robust BCL11B+ and TBR1+ deep layer cortical neurons at 2 months by immunohistochemistry. In contrast, poorly-patterned organoids contain mesendoderm-related cells, identifiable by negative QC markers including COL1A2 . Using this improved protocol, we demonstrate increased sensitivity to study the impact of different MAPT mutations from patients with frontotemporal dementia (FTD), revealing early changes in key metabolic pathways.
Collapse
|
30
|
Temple S. Advancing cell therapy for neurodegenerative diseases. Cell Stem Cell 2023; 30:512-529. [PMID: 37084729 PMCID: PMC10201979 DOI: 10.1016/j.stem.2023.03.017] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/20/2023] [Accepted: 03/28/2023] [Indexed: 04/23/2023]
Abstract
Cell-based therapies are being developed for various neurodegenerative diseases that affect the central nervous system (CNS). Concomitantly, the roles of individual cell types in neurodegenerative pathology are being uncovered by genetic and single-cell studies. With a greater understanding of cellular contributions to health and disease and with the arrival of promising approaches to modulate them, effective therapeutic cell products are now emerging. This review examines how the ability to generate diverse CNS cell types from stem cells, along with a deeper understanding of cell-type-specific functions and pathology, is advancing preclinical development of cell products for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sally Temple
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA.
| |
Collapse
|
31
|
Ampartzidis I, Efstathiou C, Paonessa F, Thompson EM, Wilson T, McCann CJ, Greene NDE, Copp AJ, Livesey FJ, Elvassore N, Giobbe GG, De Coppi P, Maniou E, Galea GL. Synchronisation of apical constriction and cell cycle progression is a conserved behaviour of pseudostratified neuroepithelia informed by their tissue geometry. Dev Biol 2023; 494:60-70. [PMID: 36509125 PMCID: PMC10570144 DOI: 10.1016/j.ydbio.2022.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/03/2022] [Accepted: 12/08/2022] [Indexed: 12/13/2022]
Abstract
Neuroepithelial cells balance tissue growth requirement with the morphogenetic imperative of closing the neural tube. They apically constrict to generate mechanical forces which elevate the neural folds, but are thought to apically dilate during mitosis. However, we previously reported that mitotic neuroepithelial cells in the mouse posterior neuropore have smaller apical surfaces than non-mitotic cells. Here, we document progressive apical enrichment of non-muscle myosin-II in mitotic, but not non-mitotic, neuroepithelial cells with smaller apical areas. Live-imaging of the chick posterior neuropore confirms apical constriction synchronised with mitosis, reaching maximal constriction by anaphase, before division and re-dilation. Mitotic apical constriction amplitude is significantly greater than interphase constrictions. To investigate conservation in humans, we characterised early stages of iPSC differentiation through dual SMAD-inhibition to robustly produce pseudostratified neuroepithelia with apically enriched actomyosin. These cultured neuroepithelial cells achieve an equivalent apical area to those in mouse embryos. iPSC-derived neuroepithelial cells have large apical areas in G2 which constrict in M phase and retain this constriction in G1/S. Given that this differentiation method produces anterior neural identities, we studied the anterior neuroepithelium of the elevating mouse mid-brain neural tube. Instead of constricting, mid-brain mitotic neuroepithelial cells have larger apical areas than interphase cells. Tissue geometry differs between the apically convex early midbrain and flat posterior neuropore. Culturing human neuroepithelia on equivalently convex surfaces prevents mitotic apical constriction. Thus, neuroepithelial cells undergo high-amplitude apical constriction synchronised with cell cycle progression but the timing of their constriction if influenced by tissue geometry.
Collapse
Affiliation(s)
- Ioakeim Ampartzidis
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Christoforos Efstathiou
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Francesco Paonessa
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK; UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research Into Rare Disease in Children, London, UK
| | - Elliott M Thompson
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Tyler Wilson
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Conor J McCann
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Nicholas DE Greene
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Andrew J Copp
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Frederick J Livesey
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK; UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research Into Rare Disease in Children, London, UK
| | - Nicola Elvassore
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK; Veneto Institute of Molecular Medicine, Padova, Italy; UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research Into Rare Disease in Children, London, UK
| | - Giovanni G Giobbe
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK; UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research Into Rare Disease in Children, London, UK
| | - Paolo De Coppi
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK; UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research Into Rare Disease in Children, London, UK; Specialist Neonatal and Paediatric Unit, Great Ormond Street Hospital, London, WC1N 1EH, UK
| | - Eirini Maniou
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK; Veneto Institute of Molecular Medicine, Padova, Italy
| | - Gabriel L Galea
- Developmental Biology and Cancer Department, UCL GOS Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK.
| |
Collapse
|
32
|
Whye D, Wood D, Saber WA, Norabuena EM, Makhortova NR, Sahin M, Buttermore ED. A Robust Pipeline for the Multi-Stage Accelerated Differentiation of Functional 3D Cortical Organoids from Human Pluripotent Stem Cells. Curr Protoc 2023; 3:e641. [PMID: 36633423 PMCID: PMC9839317 DOI: 10.1002/cpz1.641] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Disordered cellular development, abnormal neuroanatomical formations, and dysfunction of neuronal circuitry are among the pathological manifestations of cortical regions in the brain that are often implicated in complex neurodevelopmental disorders. With the advancement of stem cell methodologies such as cerebral organoid generation, it is possible to study these processes in vitro using 3D cellular platforms that mirror key developmental stages occurring throughout embryonic neurogenesis. Patterning-based stem cell models of directed neuronal development offer one approach to accomplish this, but these protocols often require protracted periods of cell culture to generate diverse cell types and current methods are plagued by a lack of specificity, reproducibility, and temporal control over cell derivation. Although ectopic expression of transcription factors offers another avenue to rapidly generate neurons, this process of direct lineage conversion bypasses critical junctures of neurodevelopment during which disease-relevant manifestations may occur. Here, we present a directed differentiation approach for generating human pluripotent stem cell (hPSC)-derived cortical organoids with accelerated lineage specification to generate functionally mature cortical neurons in a shorter timeline than previously established protocols. This novel protocol provides precise guidance for the specification of neuronal cell type identity as well as temporal control over the pace at which cortical lineage trajectories are established. Furthermore, we present assays that can be used as tools to interrogate stage-specific developmental signaling mechanisms. By recapitulating major components of embryonic neurogenesis, this protocol allows for improved in vitro modeling of cortical development while providing a platform that can be utilized to uncover disease-specific mechanisms of disordered development at various stages across the differentiation timeline. © 2023 Wiley Periodicals LLC. Basic Protocol 1: 3D hPSC neural induction Support Protocol 1: Neural rosette formation assay Support Protocol 2: Neurosphere generation Support Protocol 3: Enzymatic dissociation, NSC expansion, and cryopreservation Basic Protocol 2: 3D neural progenitor expansion Basic Protocol 3: 3D accelerated cortical lineage patterning and terminal differentiation.
Collapse
Affiliation(s)
- Dosh Whye
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
| | - Delaney Wood
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
| | - Wardiya Afshar Saber
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
- Department of Neurology, Harvard Medical School, Boston, MA
| | - Erika M. Norabuena
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
| | - Nina R. Makhortova
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
- Department of Neurology, Harvard Medical School, Boston, MA
| | - Mustafa Sahin
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
- Department of Neurology, Harvard Medical School, Boston, MA
| | - Elizabeth D. Buttermore
- Human Neuron Core, Rosamund Stone Zander Translational Neuroscience Center, Boston Children’s Hospital, Boston, MA
- F.M. Kirby Neurobiology Department, Boston Children’s Hospital, Boston, MA
| |
Collapse
|
33
|
iPSC-Derived Macrophages: The Differentiation Protocol Affects Cell Immune Characteristics and Differentiation Trajectories. Int J Mol Sci 2022; 23:ijms232416087. [PMID: 36555728 PMCID: PMC9781144 DOI: 10.3390/ijms232416087] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
The generation of human macrophages from induced pluripotent stem cells (iMacs) is a rapidly developing approach used to create disease models, screen drugs, study macrophage-pathogen interactions and develop macrophage-based cell therapy. To generate iMacs, different types of protocols have been suggested, all thought to result in the generation of similar iMac populations. However, direct comparison of iMacs generated using different protocols has not been performed. We have compared the productivity, the differentiation trajectories and the characteristics of iMacs generated using two widely used protocols: one based on the formation of embryoid bodies and the induction of myeloid differentiation by only two cytokines, interleukin-3 and macrophage colony-stimulating factor, and the other utilizing multiple exogenous factors for iMac generation. We report inter-protocol differences in the following: (i) protocol productivity; (ii) dynamic changes in the expression of genes related to inflammation and lipid homeostasis following iMac differentiation and (iii) the transcriptomic profiles of terminally differentiated iMacs, including the expression of genes involved in inflammatory response, antigen presentation and lipid homeostasis. The results document the dependence of fine iMac characteristics on the type of differentiation protocol, which is important for further development of the field, including the development of iMac-based cell therapy.
Collapse
|
34
|
Elder N, Fattahi F, McDevitt TC, Zholudeva LV. Diseased, differentiated and difficult: Strategies for improved engineering of in vitro neurological systems. Front Cell Neurosci 2022; 16:962103. [PMID: 36238834 PMCID: PMC9550918 DOI: 10.3389/fncel.2022.962103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 08/22/2022] [Indexed: 12/01/2022] Open
Abstract
The rapidly growing field of cellular engineering is enabling scientists to more effectively create in vitro models of disease and develop specific cell types that can be used to repair damaged tissue. In particular, the engineering of neurons and other components of the nervous system is at the forefront of this field. The methods used to engineer neural cells can be largely divided into systems that undergo directed differentiation through exogenous stimulation (i.e., via small molecules, arguably following developmental pathways) and those that undergo induced differentiation via protein overexpression (i.e., genetically induced and activated; arguably bypassing developmental pathways). Here, we highlight the differences between directed differentiation and induced differentiation strategies, how they can complement one another to generate specific cell phenotypes, and impacts of each strategy on downstream applications. Continued research in this nascent field will lead to the development of improved models of neurological circuits and novel treatments for those living with neurological injury and disease.
Collapse
Affiliation(s)
- Nicholas Elder
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, United States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
- Gladstone Institutes, San Francisco, CA, United States
| | - Faranak Fattahi
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, United States
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, United States
| | - Todd C. McDevitt
- Gladstone Institutes, San Francisco, CA, United States
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, United States
- Sana Biotechnology, Inc., South San Francisco, CA, United States
| | - Lyandysha V. Zholudeva
- Gladstone Institutes, San Francisco, CA, United States
- *Correspondence: Lyandysha V. Zholudeva,
| |
Collapse
|
35
|
Wang Y, Madhusudan S, Cotellessa L, Kvist J, Eskici N, Yellapragada V, Pulli K, Lund C, Vaaralahti K, Tuuri T, Giacobini P, Raivio T. Deciphering the Transcriptional Landscape of Human Pluripotent Stem Cell-Derived GnRH Neurons: The Role of Wnt Signaling in Patterning the Neural Fate. Stem Cells 2022; 40:1107-1121. [PMID: 36153707 PMCID: PMC9806769 DOI: 10.1093/stmcls/sxac069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/14/2022] [Indexed: 01/05/2023]
Abstract
Hypothalamic gonadotropin-releasing hormone (GnRH) neurons lay the foundation for human development and reproduction; however, the critical cell populations and the entangled mechanisms underlying the development of human GnRH neurons remain poorly understood. Here, by using our established human pluripotent stem cell-derived GnRH neuron model, we decoded the cellular heterogeneity and differentiation trajectories at the single-cell level. We found that a glutamatergic neuron population, which generated together with GnRH neurons, showed similar transcriptomic properties with olfactory sensory neuron and provided the migratory path for GnRH neurons. Through trajectory analysis, we identified a specific gene module activated along the GnRH neuron differentiation lineage, and we examined one of the transcription factors, DLX5, expression in human fetal GnRH neurons. Furthermore, we found that Wnt inhibition could increase DLX5 expression and improve the GnRH neuron differentiation efficiency through promoting neurogenesis and switching the differentiation fates of neural progenitors into glutamatergic neurons/GnRH neurons. Our research comprehensively reveals the dynamic cell population transition and gene regulatory network during GnRH neuron differentiation.
Collapse
Affiliation(s)
- Yafei Wang
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Shrinidhi Madhusudan
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Ludovica Cotellessa
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Jouni Kvist
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Nazli Eskici
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Venkatram Yellapragada
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kristiina Pulli
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Carina Lund
- Folkhälsan Research Center, Helsinki, Finland
| | - Kirsi Vaaralahti
- Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland,New Children’s Hospital, Pediatric Research Center, Helsinki University Hospital, Helsinki, Finland
| | - Timo Tuuri
- Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland
| | | | - Taneli Raivio
- Corresponding author: Taneli Raivio, Stem Cells and Metabolism Research Program, Research Programs Unit, and Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
36
|
Cho S, Discher DE, Leong KW, Vunjak-Novakovic G, Wu JC. Challenges and opportunities for the next generation of cardiovascular tissue engineering. Nat Methods 2022; 19:1064-1071. [PMID: 36064773 PMCID: PMC12061062 DOI: 10.1038/s41592-022-01591-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 07/07/2022] [Indexed: 12/21/2022]
Abstract
Engineered cardiac tissues derived from human induced pluripotent stem cells offer unique opportunities for patient-specific disease modeling, drug discovery and cardiac repair. Since the first engineered hearts were introduced over two decades ago, human induced pluripotent stem cell-based three-dimensional cardiac organoids and heart-on-a-chip systems have now become mainstays in basic cardiovascular research as valuable platforms for investigating fundamental human pathophysiology and development. However, major obstacles remain to be addressed before the field can truly advance toward commercial and clinical translation. Here we provide a snapshot of the state-of-the-art methods in cardiac tissue engineering, with a focus on in vitro models of the human heart. Looking ahead, we discuss major challenges and opportunities in the field and suggest strategies for enabling broad acceptance of engineered cardiac tissues as models of cardiac pathophysiology and testbeds for the development of therapies.
Collapse
Affiliation(s)
- Sangkyun Cho
- Stanford Cardiovascular Institute, Stanford, CA, USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford, CA, USA
| | - Dennis E Discher
- Molecular & Cell Biophysics Laboratory, University of Pennsylvania, Philadelphia, PA, USA
- Pennsylvania Muscle Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Medicine, Columbia University, New York, NY, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford, CA, USA.
- Department of Medicine, Division of Cardiovascular Medicine, Stanford, CA, USA.
| |
Collapse
|
37
|
de Luzy I, Pavan C, Moriarty N, Hunt C, Vandenhoven Z, Khanna A, Niclis J, Gantner C, Thompson L, Parish C. Identifying the optimal developmental age of human pluripotent stem cell-derived midbrain dopaminergic progenitors for transplantation in a rodent model of Parkinson's disease. Exp Neurol 2022; 358:114219. [DOI: 10.1016/j.expneurol.2022.114219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/15/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022]
|
38
|
Farcy S, Albert A, Gressens P, Baffet AD, El Ghouzzi V. Cortical Organoids to Model Microcephaly. Cells 2022; 11:2135. [PMID: 35883578 PMCID: PMC9320662 DOI: 10.3390/cells11142135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/17/2022] [Accepted: 07/05/2022] [Indexed: 02/01/2023] Open
Abstract
How the brain develops and achieves its final size is a fascinating issue that questions cortical evolution across species and man's place in the animal kingdom. Although animal models have so far been highly valuable in understanding the key steps of cortical development, many human specificities call for appropriate models. In particular, microcephaly, a neurodevelopmental disorder that is characterized by a smaller head circumference has been challenging to model in mice, which often do not fully recapitulate the human phenotype. The relatively recent development of brain organoid technology from induced pluripotent stem cells (iPSCs) now makes it possible to model human microcephaly, both due to genetic and environmental origins, and to generate developing cortical tissue from the patients themselves. These 3D tissues rely on iPSCs differentiation into cortical progenitors that self-organize into neuroepithelial rosettes mimicking the earliest stages of human neurogenesis in vitro. Over the last ten years, numerous protocols have been developed to control the identity of the induced brain areas, the reproducibility of the experiments and the longevity of the cultures, allowing analysis of the later stages. In this review, we describe the different approaches that instruct human iPSCs to form cortical organoids, summarize the different microcephalic conditions that have so far been modeled by organoids, and discuss the relevance of this model to decipher the cellular and molecular mechanisms of primary and secondary microcephalies.
Collapse
Affiliation(s)
- Sarah Farcy
- Institut Curie, PSL Research University, CNRS UMR144, F-75005 Paris, France;
| | - Alexandra Albert
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (A.A.); (P.G.)
| | - Pierre Gressens
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (A.A.); (P.G.)
| | - Alexandre D. Baffet
- Institut Curie, PSL Research University, CNRS UMR144, F-75005 Paris, France;
| | - Vincent El Ghouzzi
- NeuroDiderot, Inserm, Université Paris Cité, F-75019 Paris, France; (A.A.); (P.G.)
| |
Collapse
|
39
|
Cell–Cell Contact Mediates Gene Expression and Fate Choice of Human Neural Stem/Progenitor Cells. Cells 2022; 11:cells11111741. [PMID: 35681435 PMCID: PMC9179342 DOI: 10.3390/cells11111741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/21/2022] [Accepted: 05/12/2022] [Indexed: 02/04/2023] Open
Abstract
Transplantation of Neural Stem/Progenitor Cells (NPCs) is a promising regenerative strategy to promote neural repair following injury and degeneration because of the ability of these cells to proliferate, migrate, and integrate with the host tissue. Precise in vitro control of NPC proliferation without compromising multipotency and differentiation ability is critical in stem cell maintenance. This idea was highlighted in recent clinical trials, where discrepancies in NPC culturing protocols produced inconsistent therapeutic benefits. Of note, cell density plays an important role in regulating the survival, proliferation, differentiation, and fate choice of stem cells. To determine the extent of variability produced by inconsistent culturing densities, the present study cultured human-induced pluripotent NPCs (hiPSC-NPCs) at either a low or high plating density. hiPSC-NPCs were then isolated for transcriptomic analysis or differentiation in vitro. Following sequencing analysis, genes involved in cell–cell contact-mediated pathways, including Hippo-signaling, NOTCH, and WNT were differentially expressed. Modulation of these pathways was highly associated with the regulation of pro-neuronal transcription factors, which were also upregulated in response to higher-density hiPSC-NPC culture. Moreover, higher plating density translated into a greater neuronal and less astrocytic differentiation in vitro. This study highlights the importance of precisely controlling culture conditions during the development of NPC transplantation therapies.
Collapse
|
40
|
O’Hara-Wright M, Mobini S, Gonzalez-Cordero A. Bioelectric Potential in Next-Generation Organoids: Electrical Stimulation to Enhance 3D Structures of the Central Nervous System. Front Cell Dev Biol 2022; 10:901652. [PMID: 35656553 PMCID: PMC9152151 DOI: 10.3389/fcell.2022.901652] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/02/2022] [Indexed: 12/21/2022] Open
Abstract
Pluripotent stem cell-derived organoid models of the central nervous system represent one of the most exciting areas in in vitro tissue engineering. Classically, organoids of the brain, retina and spinal cord have been generated via recapitulation of in vivo developmental cues, including biochemical and biomechanical. However, a lesser studied cue, bioelectricity, has been shown to regulate central nervous system development and function. In particular, electrical stimulation of neural cells has generated some important phenotypes relating to development and differentiation. Emerging techniques in bioengineering and biomaterials utilise electrical stimulation using conductive polymers. However, state-of-the-art pluripotent stem cell technology has not yet merged with this exciting area of bioelectricity. Here, we discuss recent findings in the field of bioelectricity relating to the central nervous system, possible mechanisms, and how electrical stimulation may be utilised as a novel technique to engineer “next-generation” organoids.
Collapse
Affiliation(s)
- Michelle O’Hara-Wright
- Stem Cell Medicine Group, Children’s Medical Research Institute, University of Sydney, Westmead, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| | - Sahba Mobini
- Instituto de Micro y Nanotecnología, IMN-CNM, CSIC (CEI UAM + CSIC), Madrid, Spain
| | - Anai Gonzalez-Cordero
- Stem Cell Medicine Group, Children’s Medical Research Institute, University of Sydney, Westmead, NSW, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
- *Correspondence: Anai Gonzalez-Cordero,
| |
Collapse
|
41
|
Lee JH, Yoo G, Choi J, Park SH, Shin H, Prasad R, Lee Y, Ahn MR, Cho IJ, Sun W. Cell-line dependency in cerebral organoid induction: cautionary observations in Alzheimer’s disease patient-derived induced pluripotent stem cells. Mol Brain 2022; 15:46. [PMID: 35578344 PMCID: PMC9109296 DOI: 10.1186/s13041-022-00928-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/28/2022] [Indexed: 11/28/2022] Open
Abstract
The cerebral organoid (CO) model has been used in the study of various neurodegenerative diseases owing to its physiological implications. However, the CO model may only be representative of certain clinical findings in affected patients, while some features are not recapitulated. In this study, we found that neurons in the CO model from patients with Alzheimer’s disease were less responsive to depolarization, in contrast to previous reports. This difference may be partly attributed to the variations in brain spatial identity depending on the genetic background of the induced pluripotent stem cells. Our current observation raises concerns that the phenotypes observed in the CO model need to be carefully evaluated for their clinical implications.
Collapse
|
42
|
Sears KE, Gullapalli K, Trivedi D, Mihas A, Bukys MA, Jensen J. Controlling neural territory patterning from pluripotency using a systems developmental biology approach. iScience 2022; 25:104133. [PMID: 35434550 PMCID: PMC9010746 DOI: 10.1016/j.isci.2022.104133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 06/09/2021] [Accepted: 03/17/2022] [Indexed: 11/18/2022] Open
Abstract
Successful manufacture of specialized human cells requires process understanding of directed differentiation. Here, we apply high-dimensional Design of Experiments (HD-DoE) methodology to identify critical process parameters (CPPs) that govern neural territory patterning from pluripotency—the first stage toward specification of central nervous system (CNS) cell fates. Using computerized experimental design, 7 developmental signaling pathways were simultaneously perturbed in human pluripotent stem cell culture. Regionally specific genes spanning the anterior-posterior and dorsal-ventral axes of the developing embryo were measured after 3 days and mathematical models describing pathway control were developed using regression analysis. High-dimensional models revealed particular combinations of signaling inputs that induce expression profiles consistent with emerging CNS territories and defined CPPs for anterior and posterior neuroectoderm patterning. The results demonstrate the importance of combinatorial control during neural induction and challenge the use of generic neural induction strategies such as dual-SMAD inhibition, when seeking to specify particular lineages from pluripotency. Mathematical models describe pathway control of neuroectoderm marker expression Stage 1 media conditions optimized for regionally specific neuroectoderm in 3 days Optimized conditions are more consistent than dual-SMADi across hiPSC lines
Collapse
|
43
|
Barak M, Fedorova V, Pospisilova V, Raska J, Vochyanova S, Sedmik J, Hribkova H, Klimova H, Vanova T, Bohaciakova D. Human iPSC-Derived Neural Models for Studying Alzheimer's Disease: from Neural Stem Cells to Cerebral Organoids. Stem Cell Rev Rep 2022; 18:792-820. [PMID: 35107767 PMCID: PMC8930932 DOI: 10.1007/s12015-021-10254-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2021] [Indexed: 12/05/2022]
Abstract
During the past two decades, induced pluripotent stem cells (iPSCs) have been widely used to study mechanisms of human neural development, disease modeling, and drug discovery in vitro. Especially in the field of Alzheimer’s disease (AD), where this treatment is lacking, tremendous effort has been put into the investigation of molecular mechanisms behind this disease using induced pluripotent stem cell-based models. Numerous of these studies have found either novel regulatory mechanisms that could be exploited to develop relevant drugs for AD treatment or have already tested small molecules on in vitro cultures, directly demonstrating their effect on amelioration of AD-associated pathology. This review thus summarizes currently used differentiation strategies of induced pluripotent stem cells towards neuronal and glial cell types and cerebral organoids and their utilization in modeling AD and potential drug discovery.
Collapse
Affiliation(s)
- Martin Barak
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Veronika Fedorova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Veronika Pospisilova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Jan Raska
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Simona Vochyanova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Jiri Sedmik
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
- International Clinical Research Center, St. Anne's Faculty Hospital Brno, Brno, Czech Republic
| | - Hana Hribkova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Hana Klimova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
| | - Tereza Vanova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic
- International Clinical Research Center, St. Anne's Faculty Hospital Brno, Brno, Czech Republic
| | - Dasa Bohaciakova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University Brno, Brno, Czech Republic.
- International Clinical Research Center, St. Anne's Faculty Hospital Brno, Brno, Czech Republic.
| |
Collapse
|
44
|
Miura Y, Li MY, Revah O, Yoon SJ, Narazaki G, Pașca SP. Engineering brain assembloids to interrogate human neural circuits. Nat Protoc 2022; 17:15-35. [PMID: 34992269 DOI: 10.1038/s41596-021-00632-z] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/10/2021] [Indexed: 12/12/2022]
Abstract
The development of neural circuits involves wiring of neurons locally following their generation and migration, as well as establishing long-distance connections between brain regions. Studying these developmental processes in the human nervous system remains difficult because of limited access to tissue that can be maintained as functional over time in vitro. We have previously developed a method to convert human pluripotent stem cells into brain region-specific organoids that can be fused and integrated to form assembloids and study neuronal migration. In contrast to approaches that mix cell lineages in 2D cultures or engineer microchips, assembloids leverage self-organization to enable complex cell-cell interactions, circuit formation and maturation in long-term cultures. In this protocol, we describe approaches to model long-range neuronal connectivity in human brain assembloids. We present how to generate 3D spheroids resembling specific domains of the nervous system and then how to integrate them physically to allow axonal projections and synaptic assembly. In addition, we describe a series of assays including viral labeling and retrograde tracing, 3D live imaging of axon projection and optogenetics combined with calcium imaging and electrophysiological recordings to probe and manipulate the circuits in assembloids. The assays take 3-4 months to complete and require expertise in stem cell culture, imaging and electrophysiology. We anticipate that these approaches will be useful in deciphering human-specific aspects of neural circuit assembly and in modeling neurodevelopmental disorders with patient-derived cells.
Collapse
Affiliation(s)
- Yuki Miura
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.,Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Min-Yin Li
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.,Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Omer Revah
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.,Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Se-Jin Yoon
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA.,Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Genta Narazaki
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA
| | - Sergiu P Pașca
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA, USA. .,Stanford Brain Organogenesis, Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
45
|
Fischer S, Strobel B, Weinmann J, Gillardon F. Two engineered AAV capsid variants for efficient transduction of human cortical neurons directly converted from iPSC. J Neurosci Methods 2021; 368:109457. [PMID: 34953937 DOI: 10.1016/j.jneumeth.2021.109457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/09/2021] [Accepted: 12/19/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Recombinant adeno-associated virus (AAV) is the most widely used vector for gene therapy in clinical trials. To increase transduction efficiency and specificity, novel engineered AAV variants with modified capsid sequences are evaluated in human cell cultures and non-human primates. METHODS We tested two novel AAV capsid variants, AAV2-NNPTPSR and AAV9-NVVRSSS, in human cortical neurons, which were directly converted from human induced pluripotent stem cells and cocultured with rat primary astrocytes. RESULTS AAV2-NNPTPSR variant efficiently transduced both induced human cortical glutamatergic neurons and induced human cortical GABAergic interneurons. By contrast, AAV9-NVVRSSS variant transduced both induced human cortical neurons and cocultured rat primary astrocytes. High viral titers (1E+5 viral genomes per cell) caused a significant decrease in viability of induced human cortical neurons. Low viral titers (1E+4 viral genomes per cell) led to a significant increase in the neuronal activity marker c-Fos in transduced human neurons following treatment with a potassium channel blocker. CONCLUSIONS We identified two engineered AAV capsid variants that efficiently transduce induced human cortical neurons. The threefold higher percentage of c-Fos positive, transduced human neurons may indicate functional alterations induced by viral transduction and/or transgene expression.
Collapse
Affiliation(s)
- Sandra Fischer
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riss, Germany
| | - Benjamin Strobel
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riss, Germany
| | - Jonas Weinmann
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riss, Germany
| | - Frank Gillardon
- CNS Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach an der Riss, Germany.
| |
Collapse
|
46
|
Yeh CY, Huang WH, Chen HC, Meir YJJ. Capturing Pluripotency and Beyond. Cells 2021; 10:cells10123558. [PMID: 34944066 PMCID: PMC8700150 DOI: 10.3390/cells10123558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 11/16/2022] Open
Abstract
During the development of a multicellular organism, the specification of different cell lineages originates in a small group of pluripotent cells, the epiblasts, formed in the preimplantation embryo. The pluripotent epiblast is protected from premature differentiation until exposure to inductive cues in strictly controlled spatially and temporally organized patterns guiding fetus formation. Epiblasts cultured in vitro are embryonic stem cells (ESCs), which recapitulate the self-renewal and lineage specification properties of their endogenous counterparts. The characteristics of totipotency, although less understood than pluripotency, are becoming clearer. Recent studies have shown that a minor ESC subpopulation exhibits expanded developmental potential beyond pluripotency, displaying a characteristic reminiscent of two-cell embryo blastomeres (2CLCs). In addition, reprogramming both mouse and human ESCs in defined media can produce expanded/extended pluripotent stem cells (EPSCs) similar to but different from 2CLCs. Further, the molecular roadmaps driving the transition of various potency states have been clarified. These recent key findings will allow us to understand eutherian mammalian development by comparing the underlying differences between potency network components during development. Using the mouse as a paradigm and recent progress in human PSCs, we review the epiblast's identity acquisition during embryogenesis and their ESC counterparts regarding their pluripotent fates and beyond.
Collapse
Affiliation(s)
- Chih-Yu Yeh
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-Y.Y.); (W.-H.H.)
| | - Wei-Han Huang
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-Y.Y.); (W.-H.H.)
| | - Hung-Chi Chen
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; (C.-Y.Y.); (W.-H.H.)
- Limbal Stem Cell Laboratory, Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou 333, Taiwan
- Correspondence: (H.-C.C.); (Y.-J.J.M.)
| | - Yaa-Jyuhn James Meir
- Limbal Stem Cell Laboratory, Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou 333, Taiwan
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Correspondence: (H.-C.C.); (Y.-J.J.M.)
| |
Collapse
|
47
|
Moving Towards Induced Pluripotent Stem Cell-based Therapies with Artificial Intelligence and Machine Learning. Stem Cell Rev Rep 2021; 18:559-569. [PMID: 34843066 PMCID: PMC8930923 DOI: 10.1007/s12015-021-10302-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/13/2021] [Indexed: 10/28/2022]
Abstract
The advent of induced pluripotent stem cell (iPSC) technology, which allows to transform one cell type into another, holds the promise to produce therapeutic cells and organs on demand. Realization of this objective is contingent on the ability to demonstrate quality and safety of the cellular product for its intended use. Bottlenecks and backlogs to the clinical use of iPSCs have been fully outlined and a need has emerged for safer and standardized protocols to trigger cell reprogramming and functional differentiation. Amidst great challenges, in particular associated with lengthy culture time and laborious cell characterization, a demand for faster and more accurate methods for the validation of cell identity and function at different stages of the iPSC manufacturing process has risen. Artificial intelligence-based methods are proving helpful for these complex tasks and might revolutionize the way iPSCs are managed to create surrogate cells and organs. Here, we briefly review recent progress in artificial intelligence approaches for evaluation of iPSCs and their derivatives in experimental studies.
Collapse
|
48
|
Brambach M, Ernst A, Nolbrant S, Drouin-Ouellet J, Kirkeby A, Parmar M, Olariu V. Neural tube patterning: from a minimal model for rostrocaudal patterning toward an integrated 3D model. iScience 2021; 24:102559. [PMID: 34142058 PMCID: PMC8184516 DOI: 10.1016/j.isci.2021.102559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/16/2021] [Accepted: 05/14/2021] [Indexed: 10/27/2022] Open
Abstract
Rostrocaudal patterning of the neural tube is a defining event in vertebrate brain development. This process is driven by morphogen gradients which specify the fate of neural progenitor cells, leading to the partitioning of the tube. Although this is extensively studied experimentally, an integrated view of the genetic circuitry is lacking. Here, we present a minimal gene regulatory model for rostrocaudal patterning, whose tristable topology was determined in a data-driven way. Using this model, we identified the repression of hindbrain fate as promising strategy for the improvement of current protocols for the generation of dopaminergic neurons. Furthermore, we combined our model with an established minimal model for dorsoventral patterning on a realistic 3D neural tube and found that key features of neural tube patterning could be recapitulated. Doing so, we demonstrate how data and models from different sources can be combined to simulate complex in vivo processes.
Collapse
Affiliation(s)
- Max Brambach
- Computational Biology and Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, Lund, 223 63, Sweden
| | - Ariane Ernst
- Computational Biology and Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, Lund, 223 63, Sweden
| | - Sara Nolbrant
- Departments of Experimental Medical Science and Clinical Sciences, Wallenberg Neuroscience Center, and Lund Stem Cell Center, Lund University, 221 84 Lund, Sweden
| | | | - Agnete Kirkeby
- Departments of Experimental Medical Science and Clinical Sciences, Wallenberg Neuroscience Center, and Lund Stem Cell Center, Lund University, 221 84 Lund, Sweden
| | - Malin Parmar
- Departments of Experimental Medical Science and Clinical Sciences, Wallenberg Neuroscience Center, and Lund Stem Cell Center, Lund University, 221 84 Lund, Sweden
| | - Victor Olariu
- Computational Biology and Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, Lund, 223 63, Sweden
| |
Collapse
|
49
|
Sachana M, Willett C, Pistollato F, Bal-Price A. The potential of mechanistic information organised within the AOP framework to increase regulatory uptake of the developmental neurotoxicity (DNT) in vitro battery of assays. Reprod Toxicol 2021; 103:159-170. [PMID: 34147625 PMCID: PMC8279093 DOI: 10.1016/j.reprotox.2021.06.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 05/19/2021] [Accepted: 06/04/2021] [Indexed: 12/24/2022]
Abstract
Current in vivo DNT testing for regulatory purposes is not effective. In vitro assays anchored to key neurodevelopmental processes are available. Development of Adverse Outcome Pathways is required to increase mechanistic understanding of DNT effects. DNT Integrated Approaches to Testing and Assessment for various regulatory purposes should be developed. The OECD Guidance Document on use of in vitro DNT battery of assays is currently under development.
A major challenge in regulatory developmental neurotoxicity (DNT) assessment is lack of toxicological information for many compounds. Therefore, the Test Guidelines programme of the Organisation for Economic Cooperation and Development (OECD) took the initiative to coordinate an international collaboration between diverse stakeholders to consider integration of alternative approaches towards improving the current chemical DNT testing. During the past few years, a series of workshops was organized during which a consensus was reached that incorporation of a DNT testing battery that relies on in vitro assays anchored to key neurodevelopmental processes should be developed. These key developmental processes include neural progenitor cell proliferation, neuronal and oligodendrocyte differentiation, neural cell migration, neurite outgrowth, synaptogenesis and neuronal network formation, as well key events identified in the existing Adverse Outcome Pathways (AOPs). AOPs deliver mechanistic information on the causal links between molecular initiating event, intermediate key events and an adverse outcome of regulatory concern, providing the biological context to facilitate development of Integrated Approaches to Testing and Assessment (IATA) for various regulatory purposes. Developing IATA case studies, using mechanistic information derived from AOPs, is expected to increase scientific confidence for the use of in vitro methods within an IATA, thereby facilitating regulatory uptake. This manuscript summarizes the current state of international efforts to enhance DNT testing by using an in vitro battery of assays focusing on the role of AOPs in informing the development of IATA for different regulatory purposes, aiming to deliver an OECD guidance document on use of in vitro DNT battery of assays that include in vitro data interpretation.
Collapse
Affiliation(s)
- Magdalini Sachana
- Environment Health and Safety Division, Environment Directorate, Organisation for Economic Co-Operation and Development (OECD), 75775, Paris Cedex 16, France
| | - Catherine Willett
- Humane Society International, 1255 23rd Street NW, Washington, DC, 20037, USA
| | | | - Anna Bal-Price
- European Commission Joint Research Centre (JRC), Ispra, Italy.
| |
Collapse
|
50
|
Hofbauer P, Jahnel SM, Papai N, Giesshammer M, Deyett A, Schmidt C, Penc M, Tavernini K, Grdseloff N, Meledeth C, Ginistrelli LC, Ctortecka C, Šalic Š, Novatchkova M, Mendjan S. Cardioids reveal self-organizing principles of human cardiogenesis. Cell 2021; 184:3299-3317.e22. [PMID: 34019794 DOI: 10.1016/j.cell.2021.04.034] [Citation(s) in RCA: 264] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 02/12/2021] [Accepted: 04/19/2021] [Indexed: 12/13/2022]
Abstract
Organoids capable of forming tissue-like structures have transformed our ability to model human development and disease. With the notable exception of the human heart, lineage-specific self-organizing organoids have been reported for all major organs. Here, we established self-organizing cardioids from human pluripotent stem cells that intrinsically specify, pattern, and morph into chamber-like structures containing a cavity. Cardioid complexity can be controlled by signaling that instructs the separation of cardiomyocyte and endothelial layers and by directing epicardial spreading, inward migration, and differentiation. We find that cavity morphogenesis is governed by a mesodermal WNT-BMP signaling axis and requires its target HAND1, a transcription factor linked to developmental heart chamber defects. Upon cryoinjury, cardioids initiated a cell-type-dependent accumulation of extracellular matrix, an early hallmark of both regeneration and heart disease. Thus, human cardioids represent a powerful platform to mechanistically dissect self-organization, congenital heart defects and serve as a foundation for future translational research.
Collapse
Affiliation(s)
- Pablo Hofbauer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Stefan M Jahnel
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Nora Papai
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Magdalena Giesshammer
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Alison Deyett
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Clara Schmidt
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Mirjam Penc
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Katherina Tavernini
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Nastasja Grdseloff
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Christy Meledeth
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Lavinia Ceci Ginistrelli
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Claudia Ctortecka
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Šejla Šalic
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Maria Novatchkova
- Institute of Molecular Pathology (IMP), Vienna Biocenter 1, 1030 Vienna, Austria
| | - Sasha Mendjan
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter, Dr. Bohr Gasse 3, 1030 Vienna, Austria.
| |
Collapse
|