1
|
Kim DW, Duncan LH, Xu Z, Chang M, Sejer S, Terrillion CE, Kanold PO, Place E, Blackshaw S. Decoding gene networks controlling hypothalamic and prethalamic neuron development. Cell Rep 2025; 44:115858. [PMID: 40512619 DOI: 10.1016/j.celrep.2025.115858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/14/2025] [Accepted: 05/27/2025] [Indexed: 06/18/2025] Open
Abstract
The hypothalamus and prethalamus regulate diverse physiological and behavioral processes, yet the gene regulatory networks guiding their development remain poorly defined. Using single-cell RNA and ATAC sequencing, we profile over 660,000 cells in the developing mouse hypothalamus and prethalamus between embryonic day 11 and postnatal day 8. This resource maps key transcriptional and chromatin dynamics underlying regionalization, neurogenesis, and neuronal subtype differentiation. We identify distinct neurogenic progenitor populations and uncover gene regulatory networks controlling their spatial and temporal identity. Integration with genome-wide association study data reveals that transcription factors active in supramammillary and prethalamic lineages are associated with metabolic and neuropsychiatric traits. Cross-repressive interactions among regional transcription factors reinforce hypothalamic boundaries. Functional analysis of Dlx1/2 shows that their loss disrupts GABAergic neuron specification, leading to impaired thalamic inhibition and hyperactivity. This study provides a foundational atlas of hypothalamic and prethalamic development and highlights the importance of early gene regulatory programs in health and disease.
Collapse
Affiliation(s)
- Dong Won Kim
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark.
| | - Leighton H Duncan
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zheng Xu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Minzi Chang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sara Sejer
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Chantelle E Terrillion
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Patrick O Kanold
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elsie Place
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield S10 2TN, UK
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Wang Y, Wang Z, Mao X, Zhang H, Zhang L, Yang Y, Liu B, Li X, Luo F, Sun H. Cutting-edge technologies illuminate the neural landscape of cancer: Insights into tumor development. Cancer Lett 2025; 619:217667. [PMID: 40127813 DOI: 10.1016/j.canlet.2025.217667] [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: 01/26/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
Neurogenesis constitutes a pivotal facet of malignant tumors, wherein cancer and its therapeutic interventions possess the ability to reconfigure the nervous system, establishing a pathologic feedback loop that exacerbates tumor progression. Recent strides in high-resolution imaging, single-cell analysis, multi-omics technologies, and experimental models have opened unprecedented avenues in cancer neuroscience. This comprehensive review summarizes the latest advancements of these emerging technologies in elucidating the biological mechanisms underlying tumor initiation, invasion, metastasis, and the dynamic heterogeneity of the tumor microenvironment(TME), with a specific focus on neuron-glial-tumor interactions in glioblastoma(GBM) and other neurophilic cancers. Moreover, we innovatively propose target screening processes based on sequencing technologies and database frameworks. It rigorously evaluates ongoing clinical trial drugs and efficacy while spotlighting characteristic cells in the central and peripheral TME, consolidating cancer biomarkers pivotal for future targeted therapies and management strategies. By integrating these cutting-edge findings, this review aims to offer fresh insights into tumor-nervous system interactions, establishing a robust foundation for forthcoming clinical advancements.
Collapse
Affiliation(s)
- Yajing Wang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Centre for Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Institute for Brain Science and Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhaojun Wang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Centre for Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Institute for Brain Science and Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xinyuan Mao
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Institute for Brain Science and Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, China; The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongrui Zhang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Centre for Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Institute for Brain Science and Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lu Zhang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Centre for Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Institute for Brain Science and Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yufei Yang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Centre for Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Institute for Brain Science and Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Beibei Liu
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinxu Li
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Feiyang Luo
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Centre for Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Institute for Brain Science and Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Haitao Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Centre for Laboratory Medicine, Zhujiang Hospital and the Second Clinical Medical College, Southern Medical University, Guangzhou, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital Institute for Brain Science and Intelligence, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China.
| |
Collapse
|
3
|
Tommasini D, Yoshimatsu T, Puthussery T, Baden T, Shekhar K. Comparative transcriptomic insights into the evolution of vertebrate photoreceptor types. Curr Biol 2025; 35:2228-2239.e4. [PMID: 40250432 DOI: 10.1016/j.cub.2025.03.060] [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: 10/26/2024] [Revised: 02/20/2025] [Accepted: 03/25/2025] [Indexed: 04/20/2025]
Abstract
To explore the molecular similarities and potential evolutionary origins of vertebrate photoreceptor types, we analyzed single-cell and -nucleus transcriptomic atlases from six vertebrate species: zebrafish, chicken, lizard, opossum, ground squirrel, and human. Comparative analyses identified conserved transcriptional signatures for the five ancestral photoreceptor types: red, blue, green, and UV cones, as well as rods. We further identified and validated molecular markers of the principal and accessory members of the tetrapod double cone. Comparative transcriptomics suggests that the principal member originated from ancestral red cones, although the origin of the accessory member is less clear. The gene expression variation among cone types mirrors their spectral order (red → green → blue → UV). We find that rods are highly dissimilar to all cone types, suggesting that rods may have diverged prior to the spectral diversification of cones.
Collapse
Affiliation(s)
- Dario Tommasini
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - Takeshi Yoshimatsu
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA.
| | - Teresa Puthussery
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Vision Sciences Graduate Group, University of California, Berkeley, Berkeley, CA 94720, USA; Herbert Wertheim School of Optometry and Vision Science, University of California, Berkeley, Berkeley, CA 94720, USA.
| | - Tom Baden
- Center for Sensory Neuroscience and Computation, Sussex Neuroscience, School of Life Sciences, University of Sussex, Sussex, Brighton BN1 9QG, UK.
| | - Karthik Shekhar
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Vision Sciences Graduate Group, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA; Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
4
|
Le N, Awad S, Palazzo I, Hoang T, Blackshaw S. Viral-mediated Pou5f1 (Oct4) overexpression and inhibition of Notch signaling synergistically induce neurogenic competence in mammalian Müller glia. eLife 2025; 14:RP106450. [PMID: 40388211 PMCID: PMC12088672 DOI: 10.7554/elife.106450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2025] Open
Abstract
Retinal Müller glia in cold-blooded vertebrates can reprogram into neurogenic progenitors to replace neurons lost to injury, but mammals lack this ability. While recent studies have shown that transgenic overexpression of neurogenic bHLH factors and glial-specific disruption of NFI family transcription factors and Notch signaling induce neurogenic competence in mammalian Müller glia, induction of neurogenesis in wildtype glia has thus far proven elusive. Here, we report that viral-mediated overexpression of the pluripotency factor Pou5f1 (Oct4) induces transdifferentiation of mouse Müller glia into bipolar neurons, and synergistically stimulates glial-derived neurogenesis in parallel with Notch loss of function. Single-cell multiomic analysis shows that Pou5f1 overexpression leads to widespread changes in gene expression and chromatin accessibility, inducing activity of both the neurogenic transcription factor Rfx4 and the Yamanaka factors Sox2 and Klf4. This study demonstrates that viral-mediated overexpression of Pou5f1 induces neurogenic competence in adult mouse Müller glia, identifying mechanisms that could be used in cell-based therapies for treating retinal dystrophies.
Collapse
Affiliation(s)
- Nguyet Le
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Sherine Awad
- Department of Ophthalmology and Visual Sciences, University of Michigan School of MedicineAnn ArborUnited States
- Department of Cell and Developmental Biology, University of Michigan School of MedicineAnn ArborUnited States
- Michigan Neuroscience Institute, University of Michigan School of MedicineAnn ArborUnited States
| | - Isabella Palazzo
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Thanh Hoang
- Department of Ophthalmology and Visual Sciences, University of Michigan School of MedicineAnn ArborUnited States
- Department of Cell and Developmental Biology, University of Michigan School of MedicineAnn ArborUnited States
- Michigan Neuroscience Institute, University of Michigan School of MedicineAnn ArborUnited States
| | - Seth Blackshaw
- Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Ophthalmology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Institute for Cell Engineering, Johns Hopkins University School of MedicineBaltimoreUnited States
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
5
|
Wang J, Deng X, Jian T, Yin S, Chen L, Vergnes L, Li Z, Liu H, Lee R, Lim SY, Bahn JH, Xiao X, Zhu X, Hu G, Reue K, Liu Y, Fan G. DNA methyltransferase 1 modulates mitochondrial function through bridging m 5C RNA methylation. Mol Cell 2025; 85:1999-2016.e11. [PMID: 40328247 DOI: 10.1016/j.molcel.2025.04.019] [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: 11/02/2023] [Revised: 11/25/2024] [Accepted: 04/15/2025] [Indexed: 05/08/2025]
Abstract
DNA methyltransferase 1 (DNMT1) is an enzyme known for DNA methylation maintenance. Point mutations in its replication focus targeting sequence (RFTS) domain lead to late-onset neurodegeneration, such as autosomal dominant cerebellar ataxia-deafness and narcolepsy (ADCA-DN) disorder. Here, we demonstrated that DNMT1 has the capability to bind to mRNA transcripts and facilitate 5-methylcytosine (m5C) RNA methylation by recruiting NOP2/Sun RNA methyltransferase 2 (NSUN2). RNA m5C methylation, in turn, promotes RNA stability for those genes modulating mitochondrial function. When the DNMT1 RFTS domain is mutated in mice, it triggers aberrant DNMT1-RNA interaction and significantly elevated m5C RNA methylation and RNA stability for a portion of metabolic genes. Consequently, increased levels of metabolic RNA transcripts contribute to cumulative oxidative stress, mitochondrial dysfunction, and neurological symptoms. Collectively, our results reveal a dual role of DNMT1 in regulating both DNA and RNA methylation, which further modulates mitochondrial function, shedding light on the pathogenic mechanism of DNMT1 mutation-induced neurodegeneration.
Collapse
Affiliation(s)
- Jing Wang
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| | - Xiaoqian Deng
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Tianshen Jian
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Shanshan Yin
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Linzhi Chen
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Laurent Vergnes
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zhehao Li
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Huoyuan Liu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Ryan Lee
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sin Yee Lim
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jae Hoon Bahn
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Xinshu Xiao
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, California 90095, USA
| | - Xianmin Zhu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Ganlu Hu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China
| | - Karen Reue
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yizhi Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Guoping Fan
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai 201210, China; The Scintillon Institute, 6868 Nancy Ridge Drive, San Diego, CA 92121.
| |
Collapse
|
6
|
Chen D, Lu S, Huang K, Pearson JD, Pacal M, Peidis P, McCurdy S, Yu T, Sangwan M, Nguyen A, Monnier PP, Schramek D, Zhu L, Santamaria D, Barbacid M, Akeno N, Wikenheiser-Brokamp KA, Bremner R. Cell cycle duration determines oncogenic transformation capacity. Nature 2025; 641:1309-1318. [PMID: 40307557 PMCID: PMC12119354 DOI: 10.1038/s41586-025-08935-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 03/25/2025] [Indexed: 05/02/2025]
Abstract
Oncogenic mutations are widespread in normal human tissues1. Similarly, in murine chimeras, cells carrying an oncogenic lesion contribute normal cells to adult tissues without causing cancer2-4. How lineages that escape cancer via normal development differ from the minority that succumb is unclear. Tumours exhibit characteristic cancer hallmarks; we therefore searched for hallmarks that differentiate cancer-prone lineages from resistant lineages. Here we show that total cell cycle duration (Tc) predicts transformation susceptibility across multiple tumour types. Cancer-prone Rb- and p107-deficient retina (Rb is also known as Rb1 and p107 is also known as Rbl1) exhibited defects in apoptosis, senescence, immune surveillance, angiogenesis, DNA repair, polarity and proliferation. Perturbing the SKP2-p27-CDK2/CDK1 axis could block cancer without affecting these hallmarks. Thus, cancer requires more than the presence of its hallmarks. Notably, every tumour-suppressive mutation that we tested increased Tc, and the Tc of the cell of origin of retinoblastoma cells was half that of resistant lineages. Tc also differentiated the cell of origin in Rb-/- pituitary cancer. In lung, loss of Rb and p53 (also known as Trp53) transforms neuroendocrine cells, whereas KrasG12D or BrafV600E mutations transform alveolar type 2 cells5-7. The shortest Tc consistently identified the cell of origin, regardless of mutation timing. Thus, relative Tc is a hallmark of initiation that distinguishes cancer-prone from cancer-resistant lineages in several settings, explaining how mutated cells escape transformation without inducing apoptosis, senescence or immune surveillance.
Collapse
Affiliation(s)
- Danian Chen
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
- Department of Ophthalmology and Visual Science, University of Toronto, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Department of Ophthalmology, Research Laboratory of Ophthalmology and Vision Sciences, Eye Research Institute, West China Hospital, Sichuan University, Chengdu, China
| | - Suying Lu
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Katherine Huang
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Joel D Pearson
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
- CancerCare Manitoba Research Institute and Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Marek Pacal
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Phillipos Peidis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Sean McCurdy
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Tao Yu
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Monika Sangwan
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Angela Nguyen
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Philippe P Monnier
- Department of Ophthalmology and Visual Science, University of Toronto, Toronto, Ontario, Canada
- Donald K. Johnson Eye Institute, Krembil Research Institute, University Health Network, Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Daniel Schramek
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Liang Zhu
- Department of Developmental and Molecular Biology, The Albert Einstein Comprehensive Cancer Center and Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Ophthalmology and Visual Sciences, The Albert Einstein Comprehensive Cancer Center and Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Medicine, The Albert Einstein Comprehensive Cancer Center and Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - David Santamaria
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, Salamanca, Spain
| | - Mariano Barbacid
- Molecular Oncology Program, National Center for Cancer Research (CNIO), Madrid, Spain
| | - Nagako Akeno
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Kathryn A Wikenheiser-Brokamp
- The Perinatal Institute Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pathology and Laboratory Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Rod Bremner
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada.
- Department of Ophthalmology and Visual Science, University of Toronto, Toronto, Ontario, Canada.
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
7
|
Arrigo A, Cremona O, Aragona E, Casoni F, Consalez G, Dogru RM, Hauck SM, Antropoli A, Bianco L, Parodi MB, Bandello F, Grosche A. Müller cells trophism and pathology as the next therapeutic targets for retinal diseases. Prog Retin Eye Res 2025; 106:101357. [PMID: 40254246 DOI: 10.1016/j.preteyeres.2025.101357] [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: 02/20/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025]
Abstract
Müller cells are a crucial retinal cell type involved in multiple regulatory processes and functions that are essential for retinal health and functionality. Acting as structural and functional support for retinal neurons and photoreceptors, Müller cells produce growth factors, regulate ion and fluid homeostasis, and facilitate neuronal signaling. They play a pivotal role in retinal morphogenesis and cell differentiation, significantly contributing to macular development. Due to their radial morphology and unique cytoskeletal organization, Müller cells act as optical fibers, efficiently channeling photons directly to the photoreceptors. In response to retinal damage, Müller cells undergo specific gene expression and functional changes that serve as a first line of defense for neurons, but can also lead to unwarranted cell dysfunction, contributing to cell death and neurodegeneration. In some species, Müller cells can reactivate their developmental program, promoting retinal regeneration and plasticity-a remarkable ability that holds promising therapeutic potential if harnessed in mammals. The crucial and multifaceted roles of Müller cells-that we propose to collectively call "Müller cells trophism"-highlight the necessity of maintaining their functionality. Dysfunction of Müller cells, termed "Müller cells pathology," has been associated with a plethora of retinal diseases, including age-related macular degeneration, diabetic retinopathy, vitreomacular disorders, macular telangiectasia, and inherited retinal dystrophies. In this review, we outline how even subtle disruptions in Müller cells trophism can drive the pathological cascade of Müller cells pathology, emphasizing the need for targeted therapies to preserve retinal health and prevent disease progression.
Collapse
Affiliation(s)
- Alessandro Arrigo
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy; Eye Repair Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Ottavio Cremona
- Vita-Salute San Raffaele University, Milan, Italy; Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Emanuela Aragona
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Filippo Casoni
- Vita-Salute San Raffaele University, Milan, Italy; Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giacomo Consalez
- Vita-Salute San Raffaele University, Milan, Italy; Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rüya Merve Dogru
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, 80939, Germany
| | - Alessio Antropoli
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Bianco
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Francesco Bandello
- Ophthalmology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antje Grosche
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany.
| |
Collapse
|
8
|
Phelps PE, Ha SM, Khankan RR, Mekonnen MA, Juarez G, Ingraham Dixie KL, Chen YW, Yang X. Olfactory ensheathing cells from adult female rats are hybrid glia that promote neural repair. eLife 2025; 13:RP95629. [PMID: 40297980 PMCID: PMC12040321 DOI: 10.7554/elife.95629] [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: 04/30/2025] Open
Abstract
Olfactory ensheathing cells (OECs) are unique glial cells found in both central and peripheral nervous systems where they support continuous axonal outgrowth of olfactory sensory neurons to their targets. Previously, we reported that following severe spinal cord injury, OECs transplanted near the injury site modify the inhibitory glial scar and facilitate axon regeneration past the scar border and into the lesion. To better understand the mechanisms underlying the reparative properties of OECs, we used single-cell RNA-sequencing of OECs from adult rats to study their gene expression programs. Our analyses revealed five diverse OEC subtypes, each expressing novel marker genes and pathways indicative of progenitor, axonal regeneration, secreted molecules, or microglia-like functions. We found substantial overlap of OEC genes with those of Schwann cells, but also with microglia, astrocytes, and oligodendrocytes. We confirmed established markers on cultured OECs, and localized select top genes of OEC subtypes in olfactory bulb tissue. We also show that OECs secrete Reelin and Connective tissue growth factor, extracellular matrix molecules which are important for neural repair and axonal outgrowth. Our results support that OECs are a unique hybrid glia, some with progenitor characteristics, and that their gene expression patterns indicate functions related to wound healing, injury repair, and axonal regeneration.
Collapse
Affiliation(s)
- Patricia E Phelps
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | - Sung Min Ha
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | - Rana R Khankan
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | - Mahlet A Mekonnen
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | - Giovanni Juarez
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | | | - Yen-Wei Chen
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | - Xia Yang
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| |
Collapse
|
9
|
Ai C, Li H, Wang C, Ji Y, Wallace DC, Qian J, Zhu Y, Guan MX. Vitamin A treatment restores vision failures arising from Leber's hereditary optic neuropathy-linked mtDNA mutation. JCI Insight 2025; 10:e188962. [PMID: 40036074 PMCID: PMC12038914 DOI: 10.1172/jci.insight.188962] [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: 11/07/2024] [Accepted: 02/27/2025] [Indexed: 03/06/2025] Open
Abstract
Leber hereditary optic neuropathy (LHON) is a paradigm for mitochondrial retinopathy due to mitochondrial DNA (mtDNA) mutations. However, the mechanism underlying retinal cell-specific effects of LHON-linked mtDNA mutations remains poorly understood, and there has been no effective treatment or cure for this disorder. Using a mouse model bearing an LHON-linked ND6P25L mutation, we demonstrated that the mutation caused retinal cell-specific deficiencies, especially in retinal ganglion cells (RGCs), rods, and Müller cells. Single-cell RNA sequencing revealed cell-specific dysregulation of oxidative phosphorylation and visual signaling pathways in the mutant retina. Strikingly, ND6 mutation-induced dysfunctions caused abnormal vitamin A (VA) metabolism essential for visual function. VA supplementation remarkably alleviated retinal deficiencies, including reduced fundus lesion and retinal thickness and increased numbers of RGCs, photoreceptors, and Müller cell neurites. The restoration of visual functions with VA treatment were further evidenced by correcting dysregulations of phototransduction cascade and neurotransmitter transmission and restoring electrophysiological properties. Interestingly, VA supplementation markedly rescued the abnormal mitochondrial morphologies and functions in the mutant retina. These findings provide insight into retina-specific pathophysiology of mitochondrial retinopathy arising from VA deficiency and mitochondrial dysfunction induced by mtDNA mutation and a step toward therapeutic intervention for LHON and other mitochondrial retinopathies.
Collapse
Affiliation(s)
- Cheng Ai
- Center for Mitochondrial Biomedicine and Department of Ophthalmology, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Institute of Genetics, Zhejiang University, Hangzhou, China
- Center for Genetic Medicine, Zhejiang University International School and Institute of Medicine, Yiwu, China
| | - Huiying Li
- Institute of Genetics, Zhejiang University, Hangzhou, China
| | - Chunyan Wang
- Institute of Genetics, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanchun Ji
- Institute of Genetics, Zhejiang University, Hangzhou, China
- Division of Medical Genetics and Genomics, The Children’s Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, China
| | - Douglas C. Wallace
- Center for Mitochondrial and Epigenomic Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Junbin Qian
- Institute of Genetics, Zhejiang University, Hangzhou, China
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yimin Zhu
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Min-Xin Guan
- Center for Mitochondrial Biomedicine and Department of Ophthalmology, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
- Institute of Genetics, Zhejiang University, Hangzhou, China
- Center for Genetic Medicine, Zhejiang University International School and Institute of Medicine, Yiwu, China
- Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Zhejiang University, Hangzhou, China
| |
Collapse
|
10
|
Wu Z, Liao B, Ying J, Keung J, Zheng Z, Ahola V, Xiong W. Simultaneous cyclin D1 overexpression and p27 kip1 knockdown enable robust Müller glia cell cycle reactivation in uninjured mouse retina. eLife 2025; 13:RP100904. [PMID: 40178080 PMCID: PMC11968108 DOI: 10.7554/elife.100904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
Harnessing the regenerative potential of endogenous stem cells to restore lost neurons is a promising strategy for treating neurodegenerative disorders. Müller glia (MG), the primary glial cell type in the retina, exhibit extraordinary regenerative abilities in zebrafish, proliferating and differentiating into neurons post-injury. However, the regenerative potential of mouse MG is limited by their inherent inability to re-enter the cell cycle, constrained by high levels of the cell cycle inhibitor p27Kip1 and low levels of cyclin D1. Here, we report a method to drive robust MG proliferation by adeno-associated virus (AAV)-mediated cyclin D1 overexpression and p27Kip1 knockdown. MG proliferation induced by this dual targeting vector was self-limiting, as MG re-entered cell cycle only once. As shown by single-cell RNA-sequencing, cell cycle reactivation led to suppression of interferon signaling, activation of reactive gliosis, and downregulation of glial genes in MG. Over time, the majority of the MG daughter cells retained the glial fate, resulting in an expanded MG pool. Interestingly, about 1% MG daughter cells expressed markers for retinal interneurons, suggesting latent neurogenic potential in a small MG subset. By establishing a safe, controlled method to promote MG proliferation in vivo while preserving retinal integrity, this work provides a valuable tool for combinatorial therapies integrating neurogenic stimuli to promote neuron regeneration.
Collapse
Affiliation(s)
- Zhifei Wu
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong KongHong KongChina
| | - Baoshan Liao
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong KongHong KongChina
| | - Julia Ying
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong KongHong KongChina
| | - Jan Keung
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong KongHong KongChina
- Ming Wai Lau Centre for Reparative Medicine, Karolinska InstitutetHong KongChina
| | - Zongli Zheng
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong KongHong KongChina
- Ming Wai Lau Centre for Reparative Medicine, Karolinska InstitutetHong KongChina
| | - Virpi Ahola
- Ming Wai Lau Centre for Reparative Medicine, Karolinska InstitutetHong KongChina
- Institute of Biomedicine, University of Eastern FinlandKuopioFinland
| | - Wenjun Xiong
- Department of Biomedical Sciences and Tung Biomedical Sciences Centre, City University of Hong KongHong KongChina
| |
Collapse
|
11
|
Ou S, Hori K, Muangkram Y, Himeno Y, Tachibanaki S, Amano A. Analysis of the relationship between rod cell membrane currents and the photoreceptor component of electroretinograms using a cable model. Sci Rep 2025; 15:11211. [PMID: 40175484 PMCID: PMC11965377 DOI: 10.1038/s41598-025-95998-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/25/2025] [Indexed: 04/04/2025] Open
Abstract
This study presents a one-dimensional bidomain cable model for analyzing the relationship between rod membrane currents and rod electroretinogram (ERG) waveform components. The model incorporates the detailed structural and electrophysiological properties of rod photoreceptors by assuming the distribution of various ion currents. Simulation results indicate that the outer segment current (Iphoto) primarily influences the photoreceptor component of ERG in low-intensity light, while the transient potential notch shape called "nose," observed under high-intensity light stimulation, is mainly attributed to the Ih current in the inner segment. In addition, capacitive currents in the outer segment play a crucial role in maintaining extracellular current loops when Iphoto is inactive. These findings highlight that currents other than Iphoto, such as Ih and capacitive currents, contribute significantly to the ERG waveform, particularly under high-intensity light, as theoretically suggested by Robson et al. The model successfully reproduced the experimentally measured rod ERG waveforms and their local components, providing a foundational platform for further investigation of ERG mechanisms. This enhanced understanding could lead to improved clinical applications of ERG in the diagnosis and assessment of retinal conditions. Future work will focus on refining the ion channel distribution, incorporating additional transport mechanisms, and validating the model using a broader range of experimental data to better replicate the complex electrophysiological phenomena of rod photoreceptor cells.
Collapse
Affiliation(s)
- Shaocong Ou
- Graduate School of Life Sciences, Ritsumeikan University, Kusatsu City, Shiga, Japan
| | - Kouta Hori
- Graduate School of Life Sciences, Ritsumeikan University, Kusatsu City, Shiga, Japan
| | - Yuttamol Muangkram
- Graduate School of Life Sciences, Ritsumeikan University, Kusatsu City, Shiga, Japan
| | - Yukiko Himeno
- Graduate School of Life Sciences, Ritsumeikan University, Kusatsu City, Shiga, Japan
| | - Shuji Tachibanaki
- Division of Biology, Department of Natural Science, The Jikei University School of Medicine, Tokyo, Japan
| | - Akira Amano
- Graduate School of Life Sciences, Ritsumeikan University, Kusatsu City, Shiga, Japan.
| |
Collapse
|
12
|
Pavlou M, Probst M, Kaplan L, Filippova E, Prieve AR, Rieke F, Reh TA. AAV-mediated expression of proneural factors stimulates neurogenesis from adult Müller glia in vivo. EMBO Mol Med 2025; 17:722-746. [PMID: 40050705 PMCID: PMC11982270 DOI: 10.1038/s44321-025-00209-3] [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: 09/13/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 04/11/2025] Open
Abstract
The lack of regeneration in the human central nervous system (CNS) has major health implications. To address this, we previously used transgenic mouse models to show that neurogenesis can be stimulated in the adult mammalian retina by driving regeneration programs that other species activate following injury. Expression of specific proneural factors in adult Müller glia causes them to re-enter the cell cycle and give rise to new neurons following retinal injury. To bring this strategy closer to clinical application, we now show that neurogenesis can also be stimulated when delivering these transcription factors to Müller glia using adeno-associated viral (AAV) vectors. AAV-mediated neurogenesis phenocopies the neurogenesis we observed from transgenic animals, with different proneural factor combinations giving rise to distinct neuronal subtypes in vivo. Vector-borne neurons are morphologically, transcriptomically and physiologically similar to bipolar and amacrine/ganglion-like neurons. These results represent a key step forward in developing a cellular reprogramming approach for regenerative medicine in the CNS.
Collapse
Affiliation(s)
- Marina Pavlou
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Marlene Probst
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Lew Kaplan
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Elizaveta Filippova
- Department of Agricultural and Biological Engineering, Purdue University, Lafayette, IN, USA
| | - Aric R Prieve
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Fred Rieke
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Thomas A Reh
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA.
| |
Collapse
|
13
|
Blackshaw S, Cayouette M. Timing neural development and regeneration. Curr Opin Neurobiol 2025; 91:102976. [PMID: 40010202 DOI: 10.1016/j.conb.2025.102976] [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: 09/13/2024] [Revised: 01/15/2025] [Accepted: 01/18/2025] [Indexed: 02/28/2025]
Abstract
Regulation of neural progenitor temporal identity is critical to control the chronological order of cell birth and generation of cell diversity in the developing central nervous system (CNS). Single-cell RNA sequencing studies have identified transcriptionally distinct early and late temporal identity states in mammalian neural progenitors in multiple CNS regions. This review discusses recent advances in understanding the mechanisms underlying regulation of temporal identity in mammalian neural progenitors, the implications of these findings for glia-to-neuron reprogramming strategies, and their potential therapeutic applications. We highlight potential future directions of research, including integrating temporal identity specification with proneural factor overexpression to enhance reprogramming efficiency and broaden the repertoire of neuronal subtypes generated from reprogrammed mammalian glia.
Collapse
Affiliation(s)
- Seth Blackshaw
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 212872, USA.
| | - Michel Cayouette
- Cellular Neurobiology Research Unit, Institut de Recherches Cliniques de Montréal, Montreal, QC H2W 1R7, Canada; Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 1A1, Canada.
| |
Collapse
|
14
|
Le N, Awad S, Palazzo I, Hoang T, Blackshaw S. Viral-mediated Oct4 overexpression and inhibition of Notch signaling synergistically induce neurogenic competence in mammalian Muller glia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.09.18.613666. [PMID: 39345433 PMCID: PMC11429848 DOI: 10.1101/2024.09.18.613666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Retinal Muller glia in cold-blooded vertebrates can reprogram into neurogenic progenitors to replace neurons lost to injury, but mammals lack this ability. While recent studies have shown that transgenic overexpression of neurogenic bHLH factors and glial-specific disruption of NFI family transcription factors and Notch signaling induce neurogenic competence in mammalian Muller glia, induction of neurogenesis in wild-type glia has thus far proven elusive. Here, we report that viral-mediated overexpression of the pluripotency factor Oct4 (Pou5f1) induces transdifferentiation of mouse Muller glia into bipolar neurons, and synergistically stimulates glial-derived neurogenesis in parallel with Notch loss of function. Single cell multiomic analysis shows that Oct4 overexpression leads to widespread changes in gene expression and chromatin accessibility, inducing activity of both the neurogenic transcription factor Rfx4 and the Yamanaka factors Sox2 and Klf4. This study demonstrates that viral-mediated overexpression of Oct4 induces neurogenic competence in retinal Muller glia, identifying mechanisms that could be used in cell-based therapies for treating retinal dystrophies.
Collapse
|
15
|
Heilman SA, Schriever HC, Kostka D, Koenig KM, Gross JM. tet2 and tet3 regulate cell fate specification and differentiation events during retinal development. Sci Rep 2025; 15:10404. [PMID: 40140485 PMCID: PMC11947307 DOI: 10.1038/s41598-025-93825-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Tet family methylcytosine dioxygenases recognize and oxidize 5-methyl-cytosine (5mC) to 5-hydroxymethylcytosine (5hmC). Previous work demonstrated the requirement for Tet and 5hmC during zebrafish retinogenesis. tet2-/-;tet3-/- mutants possessed defects in the formation of differentiated retinal neurons, but the mechanisms underlying these defects are unknown. Here, we leveraged scRNAseq technologies to better understand cell type-specific deficits and molecular signatures underlying the tet2-/-;tet3-/- retinal phenotype. Our results identified defects in tet2-/-;tet3-/- retinae that included delayed specification of several retinal cell types, reduced maturity across late-stage cones, expansions of immature subpopulations of horizontal and bipolar cells, and altered biases of bipolar cell subtype fates at late differentiation stages. Together, these data highlight the critical role that tet2 and tet3 play as regulators of cell fate specification and terminal differentiation events during retinal development.
Collapse
Affiliation(s)
- Shea A Heilman
- Department of Ophthalmology, The Louis J. Fox Center for Vision Restoration, The McGowan Institute for Regenerative Medicine, The University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hannah C Schriever
- Department of Computational Biology, The University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dennis Kostka
- Department of Computational Biology, The University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kristen M Koenig
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Jeffrey M Gross
- Department of Ophthalmology, The Louis J. Fox Center for Vision Restoration, The McGowan Institute for Regenerative Medicine, The University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
16
|
Lee EJ, Kim M, Park S, Shim JH, Cho HJ, Park JA, Park K, Lee D, Kim JH, Jeong H, Matsuzaki F, Kim SY, Kim J, Yang H, Lee JS, Kim JW. Restoration of retinal regenerative potential of Müller glia by disrupting intercellular Prox1 transfer. Nat Commun 2025; 16:2928. [PMID: 40133314 PMCID: PMC11937340 DOI: 10.1038/s41467-025-58290-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
Individuals with retinal degenerative diseases struggle to restore vision due to the inability to regenerate retinal cells. Unlike cold-blooded vertebrates, mammals lack Müller glia (MG)-mediated retinal regeneration, indicating the limited regenerative capacity of mammalian MG. Here, we identify prospero-related homeobox 1 (Prox1) as a key factor restricting this process. Prox1 accumulates in MG of degenerating human and mouse retinas but not in regenerating zebrafish. In mice, Prox1 in MG originates from neighboring retinal neurons via intercellular transfer. Blocking this transfer enables MG reprogramming into retinal progenitor cells in injured mouse retinas. Moreover, adeno-associated viral delivery of an anti-Prox1 antibody, which sequesters extracellular Prox1, promotes retinal neuron regeneration and delays vision loss in a retinitis pigmentosa model. These findings establish Prox1 as a barrier to MG-mediated regeneration and highlight anti-Prox1 therapy as a promising strategy for restoring retinal regeneration in mammals.
Collapse
Affiliation(s)
- Eun Jung Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KAIST Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- Celliaz Ltd., Daejeon, South Korea
| | - Museong Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KAIST Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Sooyeon Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KAIST Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- Celliaz Ltd., Daejeon, South Korea
| | | | - Hyun-Ju Cho
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
- KRIBB School, University of Science and Technology, Daejeon, South Korea
| | | | - Kihyun Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Dongeun Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KAIST Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Jeong Hwan Kim
- Korea Bioinformation Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Haeun Jeong
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KAIST Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Fumio Matsuzaki
- Laboratory for Cell Asymmetry, RIKEN Centre for Biosystems Dynamics Research, Kobe, Hyogo, Japan
- Department of Aging Science and Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Seon-Young Kim
- Korea Bioinformation Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Jaehoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Hanseul Yang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KAIST Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Jeong-Soo Lee
- Microbiome Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
- KRIBB School, University of Science and Technology, Daejeon, South Korea
| | - Jin Woo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea.
- KAIST Stem Cell Research Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea.
- Celliaz Ltd., Daejeon, South Korea.
| |
Collapse
|
17
|
Ay A, Guillot FH, Carey AR. Case report: ocular manifestations of NFIX-associated Malan syndrome. Ophthalmic Genet 2025:1-3. [PMID: 40125923 DOI: 10.1080/13816810.2025.2482609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 02/27/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Malan syndrome, an overgrowth disorder caused by pathogenic NFIX gene variants, is characterized by macrocephaly, distinct facial features, and intellectual disability. This case highlights the associated ophthalmologic features of this rare condition. MATERIALS AND METHODS We describe a young Spanish white man with progressive vision loss in the setting of a prior clinical diagnosis of Sotos syndrome, accompanied by developmental delay and epilepsy. RESULTS Ophthalmic examination and imaging studies revealed visual acuity of 20/50 in each eye and bilateral optic atrophy. Genetic testing identified a heterozygous pathogenic NFIX variant, confirming Malan syndrome rather than NSD1-related Sotos syndrome. CONCLUSION This case underscores the importance of genetic testing in patients with syndromic features, highlighting Malan syndrome as a differential diagnosis in cases of optic atrophy with overgrowth phenotypes.
Collapse
Affiliation(s)
- Altuğ Ay
- School of Medicine, Bahçeşehir University, Istanbul, Turkey
| | - Florian H Guillot
- Division of Neuro-Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew R Carey
- Division of Neuro-Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Kim DW, Duncan LH, Xu J, Chang M, Sørensen SS, Terrillion CE, Kanold PO, Place E, Blackshaw S. Decoding Gene Networks Controlling Hypothalamic and Prethalamic Neuron Development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.10.632449. [PMID: 39829936 PMCID: PMC11741371 DOI: 10.1101/2025.01.10.632449] [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: 01/22/2025]
Abstract
Neuronal subtypes derived from the embryonic hypothalamus and prethalamus regulate many essential physiological processes, yet the gene regulatory networks controlling their development remain poorly understood. Using single-cell RNA- and ATAC-sequencing, we analyzed mouse hypothalamic and prethalamic development from embryonic day 11 to postnatal day 8, profiling 660,000 cells in total. This identified key transcriptional and chromatin dynamics driving regionalization, neurogenesis, and differentiation. This identified multiple distinct neural progenitor populations, as well as gene regulatory networks that control their spatial and temporal identities, and their terminal differentiation into major neuronal subtypes. Integrating these results with large-scale genome-wide association study data, we identified a central role for transcription factors controlling supramammillary hypothalamic development in a broad range of metabolic and cognitive traits. Recurring cross-repressive regulatory relationships were observed between transcription factors that induced prethalamic and tuberal hypothalamic identity on the one hand and mammillary and supramammillary hypothalamic identity on the other. In postnatal animals, Dlx1/2 was found to severely disrupt GABAergic neuron specification in both the hypothalamus and prethalamus, resulting in a loss of inhibition of thalamic neurons, hypersensitivity to cold, and behavioral hyperactivity. By identifying core gene regulatory networks controlling the specification and differentiation of major hypothalamic and prethalamic neuronal cell types, this study provides a roadmap for future efforts aimed at preventing and treating a broad range of homeostatic and cognitive disorders.
Collapse
Affiliation(s)
- Dong Won Kim
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Leighton H. Duncan
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jenny Xu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Minzi Chang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sara Sejer Sørensen
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Chantelle E. Terrillion
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Patrick O. Kanold
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elsie Place
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield S10 2TN, UK
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
19
|
Lahti L, Volakakis N, Gillberg L, Yaghmaeian Salmani B, Tiklová K, Kee N, Lundén-Miguel H, Werkman M, Piper M, Gronostajski R, Perlmann T. Sox9 and nuclear factor I transcription factors regulate the timing of neurogenesis and ependymal maturation in dopamine progenitors. Development 2025; 152:dev204421. [PMID: 39995267 DOI: 10.1242/dev.204421] [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: 09/26/2024] [Accepted: 02/14/2025] [Indexed: 02/26/2025]
Abstract
Correct timing of neurogenesis is crucial for generating the correct number and subtypes of glia and neurons in the embryo, and for preventing tumours and stem cell depletion in the adults. Here, we analyse how the midbrain dopamine (mDA) neuron progenitors transition into cell cycle arrest (G0) and begin to mature into ependymal cells. Comparison of mDA progenitors from different embryonic stages revealed upregulation of the genes encoding Sox9 and nuclear factor I transcription factors during development. Their conditional inactivation in the early embryonic midbrain led to delayed G0 entry and ependymal maturation in the entire midbrain ventricular zone, reduced gliogenesis and increased generation of neurons, including mDA neurons. In contrast, their inactivation in late embryogenesis did not result in mitotic re-entry, suggesting that these factors are necessary for G0 induction, but not for its maintenance. Our characterisation of adult ependymal cells by single-cell RNA sequencing and histology show that mDA-progenitor-derived cells retain several progenitor features but also secrete neuropeptides and contact neighbouring cells and blood vessels, indicating that these cells may form part of the circumventricular organ system.
Collapse
Affiliation(s)
- Laura Lahti
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Linda Gillberg
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Katarína Tiklová
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Nigel Kee
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Maarten Werkman
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Michael Piper
- The School of Biomedical Sciences and The Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Richard Gronostajski
- Genetics, Genomics & Bioinformatics Program, University at Buffalo, Buffalo, NY 14203, USA
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
- Ludwig Institute for Cancer Research, 171 77 Stockholm, Sweden
| |
Collapse
|
20
|
Taylor OB, DeGroff N, El-Hodiri HM, Gao C, Fischer AJ. Sphingosine-1-phosphate signaling regulates the ability of Müller glia to become neurogenic, proliferating progenitor-like cells. eLife 2025; 13:RP102151. [PMID: 40047533 PMCID: PMC11884796 DOI: 10.7554/elife.102151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2025] Open
Abstract
The purpose of these studies is to investigate how Sphingosine-1-phosphate (S1P) signaling regulates glial phenotype, dedifferentiation of Müller glia (MG), reprogramming into proliferating MG-derived progenitor cells (MGPCs), and neuronal differentiation of the progeny of MGPCs in the chick retina. We found that S1P-related genes are highly expressed by retinal neurons and glia, and levels of expression were dynamically regulated following retinal damage. Drug treatments that activate S1P receptor 1 (S1PR1) or increase levels of S1P suppressed the formation of MGPCs. Conversely, treatments that inhibit S1PR1 or decrease levels of S1P stimulated the formation of MGPCs. Inhibition of S1P receptors or S1P synthesis significantly enhanced the neuronal differentiation of the progeny of MGPCs. We report that S1P-related gene expression in MG is modulated by microglia and inhibition of S1P receptors or S1P synthesis partially rescues the loss of MGPC formation in damaged retinas missing microglia. Finally, we show that TGFβ/Smad3 signaling in the resting retina maintains S1PR1 expression in MG. We conclude that the S1P signaling is dynamically regulated in MG and MGPCs in the chick retina, and activation of S1P signaling depends, in part, on signals produced by reactive microglia.
Collapse
Affiliation(s)
- Olivia B Taylor
- Department of Neuroscience, College of Medicine, The Ohio State UniversityColumbusUnited States
- Neuroscience Graduate Program, The Ohio State UniversityColumbusUnited States
| | - Nicholas DeGroff
- Department of Neuroscience, College of Medicine, The Ohio State UniversityColumbusUnited States
| | - Heithem M El-Hodiri
- Department of Neuroscience, College of Medicine, The Ohio State UniversityColumbusUnited States
| | - Chengyu Gao
- Campus Chemical Instrument Center, Mass Spectrometry and Proteomics Facility, The Ohio State UniversityColumbusUnited States
| | - Andy J Fischer
- Department of Neuroscience, College of Medicine, The Ohio State UniversityColumbusUnited States
| |
Collapse
|
21
|
Oliveira-Valença VM, Roberts JM, Fernandes-Cerqueira VM, Colmerauer CH, de Toledo BC, Santos-França PL, Linden R, Martins RAP, Rocha-Martins M, Bosco A, Vetter ML, da Silveira MS. POU4F2 overexpression promotes the genesis of retinal ganglion cell-like projection neurons from late progenitors. Development 2025; 152:DEV204297. [PMID: 39946314 DOI: 10.1242/dev.204297] [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: 07/30/2024] [Accepted: 02/03/2025] [Indexed: 03/04/2025]
Abstract
Retinal ganglion cells (RGCs) are the projection neurons of the retina, and their death promotes an irreversible blindness. Several factors were described to control their genesis during retinal development. These include Atoh7, a major orchestrator of the RGC program, and downstream targets of this transcription factor, including Pou4f factors, that in turn regulate key aspects of terminal differentiation. The absence of POU4F family genes results in defects in RGC differentiation, aberrant axonal elaboration and, ultimately, RGC death. This confirms the requirement of POU4F factors for RGC development and survival, with a crucial role in regulating RGC axon outgrowth and pathfinding. Here, we have investigated in vivo whether ectopic Pou4f2 expression in late retinal progenitor cells (late RPCs) is sufficient to induce the generation of cells with RGC properties, including long-range axon projections. We show that Pou4f2 overexpression generates RGC-like cells that share morphological and transcriptional features with RGCs that are normally generated during early development and extend axonal projections up to the brain. In conclusion, these results show that POU4F2 alone is sufficient to promote the crucial properties of projection neurons that arise from retinal progenitors outside their developmental window.
Collapse
Affiliation(s)
- Viviane Medeiros Oliveira-Valença
- Laboratory for Investigation in Neuroregeneration and Development (LINDes), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Jacqueline Marie Roberts
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Vitória Melo Fernandes-Cerqueira
- Laboratory for Investigation in Neuroregeneration and Development (LINDes), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Carolina Herkenhoff Colmerauer
- Laboratory for Investigation in Neuroregeneration and Development (LINDes), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Beatriz Cardoso de Toledo
- Laboratory for Investigation in Neuroregeneration and Development (LINDes), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Pedro Lucas Santos-França
- Laboratory for Investigation in Neuroregeneration and Development (LINDes), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Rafael Linden
- Neurogenesis Lab, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Rodrigo Alves Portela Martins
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Maurício Rocha-Martins
- Laboratory for Investigation in Neuroregeneration and Development (LINDes), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| | - Alejandra Bosco
- Department of Neurobiology, University of Utah, School of Medicine, Salt Lake City, UT 84112, USA
| | - Monica Lynn Vetter
- Department of Neurobiology, University of Utah, School of Medicine, Salt Lake City, UT 84112, USA
| | - Mariana Souza da Silveira
- Laboratory for Investigation in Neuroregeneration and Development (LINDes), Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Avenida Carlos Chagas Filho 373, 21941-902 Rio de Janeiro, Brazil
| |
Collapse
|
22
|
Perez RM, Campbell J, Goswami-Sewell D, Venkatraman R, Gomez CC, Bagnetto C, Lee A, Mattos MF, Hoon M, Zuniga-Sanchez E. Ankyrins are essential for synaptic integrity of photoreceptors in the mouse outer retina. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.11.637690. [PMID: 39990488 PMCID: PMC11844522 DOI: 10.1101/2025.02.11.637690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
The mammalian visual system consists of two distinct pathways: rod- and cone-driven vision. The rod pathway is responsible for dim light vision whereas the cone pathway mediates daylight vision and color perception. The distinct processing of visual information begins at the first synapse of rod and cone photoreceptors. The unique composition and organization of the rod and cone synapse is what allows information to be parsed into the different visual pathways. Although this is a critical process for vision, little is known about the key molecules responsible for establishing and maintaining the distinct synaptic architecture of the rod and cone synapse. In the present study, we uncovered a new role for Ankyrins in maintaining the synaptic integrity of the rod and cone synapse. Loss of Ankyrin-B and Ankyrin-G results in connectivity defects between photoreceptors and their synaptic partners. Ultrastructure analysis of the rod and cone synapse revealed impaired synaptic innervation, abnormal terminal morphology, and disruption of synaptic connections. Consistent with these findings, functional studies revealed impaired in vivo retinal responses in animals with loss of Ankyrin-B and Ankyrin-G. Taken together, our data supports a new role for Ankyrins in maintaining synaptic integrity and organization of photoreceptor synapses in the mouse outer retina. SIGNFICANCE STATEMENT The first synapse in the outer retina begins to process visual information into two distinct pathways. This is largely attributed to the different composition and organization of the rod and cone synapse. Although the structural integrity of the rod and cone synapse is critical for normal vision, little is known about the key molecules responsible for maintaining the unique structure of the different photoreceptor synapses. In this study, we demonstrate a new function for the cytoskeletal scaffolding proteins, Ankryin-B and Ankyrin-G in the mouse outer retina. We found Ankyrin-B and Ankyrin-G are both required for proper retinal connectivity, where loss of these molecules leads to synaptic defects and impaired retinal responses.
Collapse
|
23
|
Vidal-Vázquez N, Hernández-Núñez I, Carballo-Pacoret P, Salisbury S, Villamayor PR, Hervas-Sotomayor F, Yuan X, Lamanna F, Schneider C, Schmidt J, Mazan S, Kaessmann H, Adrio F, Robledo D, Barreiro-Iglesias A, Candal E. A single-nucleus RNA sequencing atlas of the postnatal retina of the shark Scyliorhinus canicula. Sci Data 2025; 12:228. [PMID: 39920165 PMCID: PMC11806052 DOI: 10.1038/s41597-025-04547-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 01/28/2025] [Indexed: 02/09/2025] Open
Abstract
The retina, whose basic cellular structure is highly conserved across vertebrates, constitutes an accessible system for studying the central nervous system. In recent years, single-cell RNA sequencing studies have uncovered cellular diversity in the retina of a variety of species, providing new insights on retinal evolution and development. However, similar data in cartilaginous fishes, the sister group to all other extant jawed vertebrates, are still lacking. Here, we present a single-nucleus RNA sequencing atlas of the postnatal retina of the catshark Scyliorhinus canicula, consisting of the expression profiles for 17,438 individual cells from three female, juvenile catshark specimens. Unsupervised clustering revealed 22 distinct cell types comprising all major retinal cell classes, as well as retinal progenitor cells (whose presence reflects the persistence of proliferative activity in postnatal stages in sharks) and oligodendrocytes. Thus, our dataset serves as a foundation for further studies on the development and function of the catshark retina. Moreover, integration of our atlas with data from other species will allow for a better understanding of vertebrate retinal evolution.
Collapse
Affiliation(s)
- Nicolás Vidal-Vázquez
- Departamento de Bioloxía Funcional, Facultade de Bioloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Aquatic One Health Research Center (ARCUS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Ismael Hernández-Núñez
- Departamento de Bioloxía Funcional, Facultade de Bioloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Pablo Carballo-Pacoret
- Departamento de Bioloxía Funcional, Facultade de Bioloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Sarah Salisbury
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, UK
| | - Paula R Villamayor
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, UK
- Departamento de Zooloxía, Xenética e Antropoloxía Física, Facultade de Veterinaria, Universidade de Santiago de Compostela, 27002, Lugo, Spain
| | - Francisca Hervas-Sotomayor
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Heidelberg, Germany
- INRAE, LPGP, Rennes, France
| | - Xuefei Yuan
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Heidelberg, Germany
| | - Francesco Lamanna
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Heidelberg, Germany
| | - Céline Schneider
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Heidelberg, Germany
| | - Julia Schmidt
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Heidelberg, Germany
| | - Sylvie Mazan
- CNRS, Sorbonne Université, Biologie Intégrative des Organismes Marins, UMR7232-BIOM, Banyuls-sur-Mer, France
| | - Henrik Kaessmann
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, Heidelberg, Germany
| | - Fátima Adrio
- Departamento de Bioloxía Funcional, Facultade de Bioloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Diego Robledo
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, EH25 9RG, UK
- Departamento de Zooloxía, Xenética e Antropoloxía Física, CIBUS, Facultade de Bioloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Antón Barreiro-Iglesias
- Departamento de Bioloxía Funcional, Facultade de Bioloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
- Aquatic One Health Research Center (ARCUS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Eva Candal
- Departamento de Bioloxía Funcional, Facultade de Bioloxía, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Aquatic One Health Research Center (ARCUS), Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain.
| |
Collapse
|
24
|
Jackson VE, Wu Y, Bonelli R, Owen JP, Scott LW, Farashi S, Kihara Y, Gantner ML, Egan C, Williams KM, Ansell BRE, Tufail A, Lee AY, Bahlo M. Multi-omic spatial effects on high-resolution AI-derived retinal thickness. Nat Commun 2025; 16:1317. [PMID: 39904976 PMCID: PMC11794613 DOI: 10.1038/s41467-024-55635-7] [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: 01/09/2024] [Accepted: 12/18/2024] [Indexed: 02/06/2025] Open
Abstract
Retinal thickness is a marker of retinal health and more broadly, is seen as a promising biomarker for many systemic diseases. Retinal thickness measurements are procured from optical coherence tomography (OCT) as part of routine clinical eyecare. We processed the UK Biobank OCT images using a convolutional neural network to produce fine-scale retinal thickness measurements across > 29,000 points in the macula, the part of the retina responsible for human central vision. The macula is disproportionately affected by high disease burden retinal disorders such as age-related macular degeneration and diabetic retinopathy, which both involve metabolic dysregulation. Analysis of common genomic variants, metabolomic, blood and immune biomarkers, disease PheCodes and genetic scores across a fine-scale macular thickness grid, reveals multiple novel genetic loci including four on the X chromosome; retinal thinning associated with many systemic disorders including multiple sclerosis; and multiple associations to correlated metabolites that cluster spatially in the retina. We highlight parafoveal thickness to be particularly susceptible to systemic insults. These results demonstrate the gains in discovery power and resolution achievable with AI-leveraged analysis. Results are accessible using a bespoke web interface that gives full control to pursue findings.
Collapse
Affiliation(s)
- V E Jackson
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Y Wu
- Department of Ophthalmology, Roger and Angie Karalis Johnson Retina Center, University of Washington, Seattle, WA, USA
| | - R Bonelli
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
- Lowy Medical Research Institute, La Jolla, CA, USA
| | - J P Owen
- Department of Ophthalmology, Roger and Angie Karalis Johnson Retina Center, University of Washington, Seattle, WA, USA
| | - L W Scott
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - S Farashi
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Y Kihara
- Department of Ophthalmology, Roger and Angie Karalis Johnson Retina Center, University of Washington, Seattle, WA, USA
| | - M L Gantner
- Lowy Medical Research Institute, La Jolla, CA, USA
| | - C Egan
- Moorfields Eye Hospital NHS Foundation Trust, London, UK
- Institute of Ophthalmology, University College London, London, UK
| | - K M Williams
- Moorfields Eye Hospital NHS Foundation Trust, London, UK
- Institute of Ophthalmology, University College London, London, UK
- Section of Ophthalmology, King's College London, London, UK
| | - B R E Ansell
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - A Tufail
- Moorfields Eye Hospital NHS Foundation Trust, London, UK
- Institute of Ophthalmology, University College London, London, UK
| | - A Y Lee
- Department of Ophthalmology, Roger and Angie Karalis Johnson Retina Center, University of Washington, Seattle, WA, USA
| | - M Bahlo
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
25
|
Hernández-Núñez I, Urman A, Zhang X, Jacobs W, Hoffman C, Rebba S, Harding EG, Li Q, Mao F, Cani AK, Chen S, Dawlaty MM, Rao RC, Ruzycki PA, Edwards JR, Clark BS. Active DNA demethylation is upstream of rod-photoreceptor fate determination and required for retinal development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.03.636318. [PMID: 39975078 PMCID: PMC11838574 DOI: 10.1101/2025.02.03.636318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Retinal cell fate specification from multipotent retinal progenitors is governed by dynamic changes in chromatin structure and gene expression. Methylation at cytosines in DNA (5mC) is actively regulated for proper control of gene expression and chromatin architecture. Numerous genes display active DNA demethylation across retinal development; a process that requires oxidation of 5mC to 5-hydroxymethylcytosine (5hmC) and is controlled by the ten-eleven translocation methylcytosine dioxygenase (TET) enzymes. Using an allelic series of conditional TET enzyme mutants, we determine that DNA demethylation is required upstream of NRL and NR2E3 expression for the establishment of rod-photoreceptor fate. Using histological, behavioral, transcriptomic, and base-pair resolution DNA methylation analyses, we establish that inhibition of active DNA demethylation results in global changes in gene expression and methylation patterns that prevent photoreceptor precursors from adopting a rod-photoreceptor fate, instead producing a retina in which all photoreceptors specify as cones. Our results establish the TET enzymes and DNA demethylation as critical regulators of retinal development and cell fate specification, elucidating a novel mechanism required for the specification of rod-photoreceptors.
Collapse
Affiliation(s)
- Ismael Hernández-Núñez
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Alaina Urman
- Center for Pharmacogenetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaodong Zhang
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - William Jacobs
- Center for Pharmacogenetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Christy Hoffman
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Sohini Rebba
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Ellen G Harding
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Qiang Li
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA
| | - Fengbiao Mao
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Andi K Cani
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Shiming Chen
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Meelad M Dawlaty
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Department of Genetics, and Department of Developmental & Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rajesh C Rao
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Department and Center of Computational Medicine and Bioinformatics, Comprehensive Cancer Center, A. Alfred Taubman Medical Research Institute, Center for RNA Biomedicine, Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
- Division of Ophthalmology, Surgery Section, VA Ann Arbor Health System, Ann Arbor, MI, USA
| | - Philip A Ruzycki
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - John R Edwards
- Center for Pharmacogenetics, Washington University School of Medicine, St. Louis, MO, USA
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian S Clark
- John F. Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
26
|
Perumal N, Gopalakrishnan P, Burkovetskaya M, Doss D, Dukkipati SS, Kanchan RK, Mahapatra S. Nuclear factor I/B: Duality in action in cancer pathophysiology. Cancer Lett 2025; 609:217349. [PMID: 39581218 DOI: 10.1016/j.canlet.2024.217349] [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: 07/07/2024] [Revised: 11/15/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
The nuclear factor I (NFI) family of transcription factors plays a decisive role in organ development and maturation. Their deregulation has been linked with various diseases, most notably cancer. NFIB stands apart from the other NFI family members given its unique ability to drive both tumor suppressive and oncogenic programs. Thus, the ultimate impact of deregulated NFIB signaling is cancer-specific and strongly influenced by an intricate network of upstream regulators and downstream effectors. Deciphering the events that drive NFIB's paradoxical roles within these networks will enable us to not only understand how this critical transcription factor enacts its dual roles but also drive innovations to help us effectively target NFIB in different cancers. Here, we provide an in-depth review of NFIB. Starting with its defining role in the development of various organs, most notably the central nervous system, we highlight critical signaling pathways and the impact of deregulation on neoplastic transformation, contrasting it with the effect of silencing alone. We then provide examples of its dual roles in various cancers, identifying specific signaling networks associated with oncogenesis versus tumor suppression. We incorporate an example of a cancer type, osteosarcoma, wherein NFIB enacts its dual functions and explore which pathways influence each function. In this manner, we suggest plausible mechanisms for its role-switching from cancers sharing common triggering events in the setting of NFIB deregulation. We also review how NFIB enhances aggressiveness by driving metastasis, stemness, and chemoresistance. We conclude with a discussion on efficacious ways to target NFIB and pose some unanswered questions that may further help solidify our understanding of NFIB and facilitate clinical translation of NFIB targeting.
Collapse
Affiliation(s)
- Naveenkumar Perumal
- School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Prakadeeswari Gopalakrishnan
- Department of Ophthalmology, Center for Translational Vision Research, Gavin Herbert Eye Institute, University of California, Irvine, CA, USA
| | - Maria Burkovetskaya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - David Doss
- School of Medicine, Creighton University, Omaha, NE, USA
| | - S Shekar Dukkipati
- Department of Pediatrics, Columbia University Irving Medical Center, New York City, NY, USA
| | - Ranjana K Kanchan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sidharth Mahapatra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
27
|
Taylor O, DeGroff N, El-Hodiri H, Gao C, Fischer AJ. Sphingosine-1-phosphate signaling regulates the ability of Müller glia to become neurogenic, proliferating progenitor-like cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.06.606815. [PMID: 39149287 PMCID: PMC11326190 DOI: 10.1101/2024.08.06.606815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
The purpose of these studies is to investigate how Sphingosine-1-phosphate (S1P) signaling regulates glial phenotype, dedifferentiation of Müller glia (MG), reprogramming into proliferating MG-derived progenitor cells (MGPCs), and neuronal differentiation of the progeny of MGPCs in the chick retina. We found that S1P-related genes are highly expressed by retinal neurons and glia, and levels of expression were dynamically regulated following retinal damage. Drug treatments that activate S1P receptor 1 (S1PR1) or increase levels of S1P suppressed the formation of MGPCs. Conversely, treatments that inhibit S1PR1 or decrease levels of S1P stimulated the formation of MGPCs. Inhibition of S1P receptors or S1P synthesis significantly enhanced the neuronal differentiation of the progeny of MGPCs. We report that S1P-related gene expression in MG is modulated by microglia and inhibition of S1P receptors or S1P synthesis partially rescues the loss of MGPC formation in damaged retinas missing microglia. Finally, we show that TGFβ/Smad3 signaling in the resting retina maintains S1PR1 expression in MG. We conclude that the S1P signaling is dynamically regulated in MG and MGPCs in the chick retina, and activation of S1P signaling depends, in part, on signals produced by reactive microglia.
Collapse
Affiliation(s)
- Olivia Taylor
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Neuroscience Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Nick DeGroff
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Heithem El-Hodiri
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Chengyu Gao
- Campus Chemical Instrument Center, Mass Spectrometry & Proteomics Facility, The Ohio State University, Columbus, OH 43210, USA
| | - Andy J. Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
28
|
Yang T, Zhang N, Yang N. Single-cell sequencing in diabetic retinopathy: progress and prospects. J Transl Med 2025; 23:49. [PMID: 39806376 PMCID: PMC11727737 DOI: 10.1186/s12967-024-06066-x] [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: 10/11/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Diabetic retinopathy is a major ocular complication of diabetes, characterized by progressive retinal microvascular damage and significant visual impairment in working-age adults. Traditional bulk RNA sequencing offers overall gene expression profiles but does not account for cellular heterogeneity. Single-cell RNA sequencing overcomes this limitation by providing transcriptomic data at the individual cell level and distinguishing novel cell subtypes, developmental trajectories, and intercellular communications. Researchers can use single-cell sequencing to draw retinal cell atlases and identify the transcriptomic features of retinal cells, enhancing our understanding of the pathogenesis and pathological changes in diabetic retinopathy. Additionally, single-cell sequencing is widely employed to analyze retinal organoids and single extracellular vesicles. Single-cell multi-omics sequencing integrates omics information, whereas stereo-sequencing analyzes gene expression and spatiotemporal data simultaneously. This review discusses the protocols of single-cell sequencing for obtaining single cells from retina and accurate sequencing data. It highlights the applications and advancements of single-cell sequencing in the study of normal retinas and the pathological changes associated with diabetic retinopathy. This underscores the potential of these technologies to deepen our understanding of the pathogenesis of diabetic retinopathy that may lead to the introduction of new therapeutic strategies.
Collapse
Affiliation(s)
- Tianshu Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, 430060, China
| | - Ningzhi Zhang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, 430060, China
| | - Ning Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Jiefang Road, Wuhan, Hubei, 430060, China.
| |
Collapse
|
29
|
Zhang Y, Ma Y, Ji YK, Jiang YF, Li D, Mu W, Yao MD, Yao J, Yan B. Co-targeting of glial activation and inflammation by tsRNA-Gln-i-0095 for treating retinal ischemic pathologies. Cell Commun Signal 2025; 23:18. [PMID: 39794828 PMCID: PMC11721595 DOI: 10.1186/s12964-024-02013-x] [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: 08/23/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
Ischemic retinopathies are the major causes of blindness, yet effective early-stage treatments remain limited due to an incomplete understanding of the underlying molecular mechanisms. Significant changes in gene expression often precede structural and functional alterations. Transfer RNA (tRNA)-derived small RNAs (tsRNAs) are emerging as novel gene regulators, involved in various biological processes and human diseases. In this study, tsRNA-Gln-i-0095 was identified as a novel regulator, which was significantly upregulated in retinal ischemia/reperfusion (I/R) injury. Reducing the levels of tsRNA-Gln-i-0095 suppressed reactive gliosis, lowered inflammatory cytokine levels, and protected retinal ganglion cells from I/R injury. These effects led to reduced structural and functional damage, inhibited glial activation and inflammation, and enhanced neuronal function. Mechanistically, tsRNA-Gln-i-0095 downregulated the expression of NFIA and TGFBR2 through a miRNA-like mechanism. Collectively, this study highlights the potential of targeting tsRNA-Gln-i-0095 as a novel therapeutic approach to reduce retinal I/R injury and preserve visual function.
Collapse
Affiliation(s)
- Ying Zhang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 210000, China
- Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Yan Ma
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 210000, China
| | - Yu-Ke Ji
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 210000, China
| | - Yi-Fei Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 210000, China
| | - Duo Li
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 210000, China
| | - Wan Mu
- Eye Institute, Department of Ophthalmology, Eye and ENT Hospital, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200030, China
| | - Mu-Di Yao
- Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200080, China.
| | - Jin Yao
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210000, China.
- The Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, 210000, China.
| | - Biao Yan
- Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200080, China.
| |
Collapse
|
30
|
Lin CH, Wu MR, Tanasa B, Prakhar P, Deng B, Davis AE, Li L, Xia A, Shan Y, Fort PE, Wang S. Induction of a Müller Glial Cell-Specific Protective Pathway Safeguards the Retina From Diabetes-Induced Damage. Diabetes 2025; 74:96-107. [PMID: 39446557 DOI: 10.2337/db24-0199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 10/19/2024] [Indexed: 10/26/2024]
Abstract
Diabetes can lead to cell type-specific responses in the retina, including vascular lesions, glial dysfunction, and neurodegeneration, all of which contribute to retinopathy. However, the molecular mechanisms underlying these cell type-specific responses, and the cell types that are sensitive to diabetes have not been fully elucidated. Using single-cell transcriptomics, we profiled the transcriptional changes induced by diabetes in different retinal cell types in rat models as the disease progressed. Rod photoreceptors, a subtype of amacrine interneurons, and Müller glial cells (MGs) exhibited rapid responses to diabetes at the transcript levels. Genes associated with ion regulation were upregulated in all three cell types, suggesting a common response to diabetes. Furthermore, focused studies revealed that although MG initially increased the expression of genes playing protective roles, they cannot sustain this beneficial effect. We explored one of the candidate protective genes, Zinc finger protein 36 homolog (Zfp36), and observed that depleting Zfp36 in rat MGs in vivo using adeno-associated virus-based tools exacerbated diabetes-induced phenotypes, including glial reactivation, neurodegeneration, and vascular defects. Overexpression of Zfp36 slowed the development of these phenotypes. This work unveiled retinal cell types that are sensitive to diabetes and demonstrated that MGs can mount protective responses through Zfp36. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Cheng-Hui Lin
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA
| | - Man-Ru Wu
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA
| | - Bogdan Tanasa
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA
| | - Praveen Prakhar
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA
| | - Boxiong Deng
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA
| | - Alexander E Davis
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA
| | - Liang Li
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA
| | - Alexander Xia
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA
| | - Yang Shan
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI
| | - Patrice E Fort
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI
| | - Sui Wang
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA
| |
Collapse
|
31
|
Hernández-Núñez I, Clark BS. Experimental Framework for Assessing Mouse Retinal Regeneration Through Single-Cell RNA-Sequencing. Methods Mol Biol 2025; 2848:117-134. [PMID: 39240520 DOI: 10.1007/978-1-0716-4087-6_8] [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: 09/07/2024]
Abstract
Retinal degenerative diseases including age-related macular degeneration and glaucoma are estimated to currently affect more than 14 million people in the United States, with an increased prevalence of retinal degenerations in aged individuals. An expanding aged population who are living longer forecasts an increased prevalence and economic burden of visual impairments. Improvements to visual health and treatment paradigms for progressive retinal degenerations slow vision loss. However, current treatments fail to remedy the root cause of visual impairments caused by retinal degenerations-loss of retinal neurons. Stimulation of retinal regeneration from endogenous cellular sources presents an exciting treatment avenue for replacement of lost retinal cells. In multiple species including zebrafish and Xenopus, Müller glial cells maintain a highly efficient regenerative ability to reconstitute lost cells throughout the organism's lifespan, highlighting potential therapeutic avenues for stimulation of retinal regeneration in humans. Here, we describe how the application of single-cell RNA-sequencing (scRNA-seq) has enhanced our understanding of Müller glial cell-derived retinal regeneration, including the characterization of gene regulatory networks that facilitate/inhibit regenerative responses. Additionally, we provide a validated experimental framework for cellular preparation of mouse retinal cells as input into scRNA-seq experiments, including insights into experimental design and analyses of resulting data.
Collapse
Affiliation(s)
- Ismael Hernández-Núñez
- John F Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian S Clark
- John F Hardesty, MD Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
32
|
Chen D, Xin Y, Guo J, Chen S. Mettl14 and Mettl3 Work Cooperatively to Regulate Retinal Development. Cell Biochem Funct 2025; 43:e70039. [PMID: 39739431 DOI: 10.1002/cbf.70039] [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: 08/03/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025]
Abstract
N6-methylenadenosine (m6A) modification, the most abundant epitranscriptomic modification in eukaryotic mRNAs, has been shown to play crucial roles in regulating various aspects of mRNA metabolism and functions. In this study, we applied the Cre-Loxp conditional knockout system to investigate the role of the core components of the m6A methyltransferase complex, METTL14 and METTL3, in retinal development. Our results showed that the double absence of Mettl14 and Mettl3 caused structural disturbance in the retina and prolonged the proliferation activity of retinal progenitor cells. Interestingly, the deletion of Mettl14 and Mettl3 did not affect the generation of various retinal cells, but severely disrupted their distribution. In addition, double deletion of Mettl14 together with Mettl3 caused a stronger phenotype than did single deletion of Mettl14. In conclusion, our study demonstrated that Mettl14 and Mettl3 work cooperatively to regulate retinal development.
Collapse
Affiliation(s)
- Dan Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Yanling Xin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Jingyi Guo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Shuyi Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| |
Collapse
|
33
|
Dillon NR, Doe CQ. Castor is a temporal transcription factor that specifies early born central complex neuron identity. Development 2024; 151:dev204318. [PMID: 39620972 DOI: 10.1242/dev.204318] [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: 08/09/2024] [Accepted: 11/20/2024] [Indexed: 12/17/2024]
Abstract
The generation of neuronal diversity is important for brain function, but how diversity is generated is incompletely understood. We used the development of the Drosophila central complex (CX) to address this question. The CX develops from eight bilateral Type 2 neuroblasts (T2NBs), which generate hundreds of different neuronal types. T2NBs express broad opposing temporal gradients of RNA-binding proteins. It remains unknown whether these protein gradients are sufficient to directly generate all known neuronal diversity, or whether there are temporal transcription factors (TTFs) with narrow expression windows that each specify a small subset of CX neuron identities. Multiple candidate TTFs have been identified, but their function remains uncharacterized. Here, we show that: (1) the adult E-PG neurons are born from early larval T2NBs; (2) the candidate TTF Castor is expressed transiently in early larval T2NBs when E-PG and P-EN neurons are born; and (3) Castor is required to specify early born E-PG and P-EN neuron identities. We conclude that Castor is a TTF in larval T2NB lineages that specifies multiple, early born CX neuron identities.
Collapse
Affiliation(s)
- Noah R Dillon
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403, USA
| | - Chris Q Doe
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403, USA
| |
Collapse
|
34
|
Schaeffer J, Belin S. Axon regeneration: an issue of translation. C R Biol 2024; 347:249-258. [PMID: 39665232 DOI: 10.5802/crbiol.169] [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: 09/27/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 12/13/2024]
Abstract
In the mammalian central nervous system (CNS), adult neurons fail to regenerate spontaneously upon axon injury, which leads to a permanent and irreversible loss of neuronal functions. For more than 15 years, much effort was invested to unlock axon regrowth programs based on extensive transcriptomic characterization. However, it is now well described that mRNA and protein levels correlate only partially in cells, and that the transcription process (from DNA to mRNA) may not directly reflect protein expression. Conversely, the translation process (from mRNA to protein) provides an additional layer of gene regulation. This aspect has been overlooked in CNS regeneration. In this review, we discuss the limitations of transcriptomic approaches to promote CNS regeneration and we provide the rationale to investigate translational regulation in this context, and notably the regulatory role of the translational complex. Finally, we summarize our and others’ recent findings showing how variations in the translational complex composition regulate selective (mRNA-specific) translation, thereby controlling CNS axon regrowth.
Collapse
|
35
|
Li T, Ma Y, Cheng Y, Zhao Y, Qiu Z, Liu H, Zhang D, Wu J, Li J, Zhang S, Wu J. Single-Cell Transcriptomic Dataset of RPGR-associated Retinitis Pigmentosa Patient-Derived Retinal Organoids. Sci Data 2024; 11:1285. [PMID: 39592612 PMCID: PMC11599861 DOI: 10.1038/s41597-024-04124-z] [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: 07/23/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
X-linked retinitis pigmentosa (XLRP) is a severe hereditary retinal disorder marked by progressive vision loss due to photoreceptor dysfunction. The retinitis pigmentosa GTPase regulator (RPGR) gene, responsible for most XLRP cases, encodes a protein crucial for the transport of visual signal proteins between the photoreceptor inner and outer segments. However, the mechanism of RPGR mutation causing photoreceptor disorder is not clear and effective treatments remain elusive. This study utilized retinal organoids (ROs) derived from normal and RPGR-mutant human induced pluripotent stem cells (hiPSC) at four developmental stages (40, 90, 150, and 200 days). Single-cell RNA sequencing (scRNA-seq) was conducted on 71,096 cells, including 33,839 cells from the control group and 37,257 cells from the RPGR group. Key retinal cell types were identified and the obtained scRNAseq dataset was validated reliable and high -quality. This study has provided data resources and references for exploring the mechanism of RPGR-related retinal degeneration and support the development of targeted therapies.
Collapse
Affiliation(s)
- Ting Li
- Qingdao Institute, College of Medicine, Fudan University, Qingdao, 266500, China
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China
| | - Yuting Ma
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- BGI Genomics, Shenzhen, 518083, China
| | - Yun Cheng
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China
| | - Yingke Zhao
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China
| | - Zhixu Qiu
- BGI Genomics, Shenzhen, 518083, China
| | - Hongli Liu
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China
| | - Daowei Zhang
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China
| | - Jiawen Wu
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China
| | - Junfeng Li
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China
| | - Shenghai Zhang
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China.
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China.
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China.
| | - Jihong Wu
- Qingdao Institute, College of Medicine, Fudan University, Qingdao, 266500, China.
- Department of Ophthalmology, Eye and ENT Hospital, College of Medicine, Fudan University, Shanghai, 200000, China.
- Shanghai Key Laboratory of Visual Impairment and Restoration, Science and Technology Commission of Shanghai Municipality, Shanghai, 200000, China.
- Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, National Health Commission, Shanghai, 200000, China.
| |
Collapse
|
36
|
Howard L, Ishikawa Y, Katayama T, Park SJ, Hill MJ, Blake DJ, Nishida K, Hayashi R, Quantock AJ. Single-cell transcriptomics reveals the molecular basis of human iPS cell differentiation into ectodermal ocular lineages. Commun Biol 2024; 7:1495. [PMID: 39532995 PMCID: PMC11557866 DOI: 10.1038/s42003-024-07130-4] [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: 04/20/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
The generation of a self-formed, ectodermal, autonomous multi-zone (SEAM) from human induced pluripotent stem cells (hiPSCs) offers a unique perspective to study the dynamics of ocular cell differentiation over time. Here, by utilising single-cell transcriptomics, we have (i) identified, (ii) molecularly characterised and (iii) ascertained the developmental trajectories of ectodermally-derived ocular cell populations which emerge within SEAMs as they form. Our analysis reveals interdependency between tissues of the early eye and delineates the sequential formation and maturation of distinct cell types over a 12-week period. We demonstrate a progression from pluripotency through to tissue specification and differentiation which encompasses both surface ectodermal and neuroectodermal ocular lineages and the generation of iPSC-derived components of the developing cornea, conjunctiva, lens, and retina. Our findings not only advance the understanding of ocular development in a stem cell-based system of human origin, but also establish a robust methodological paradigm for exploring cellular and molecular dynamics during SEAM formation at single-cell resolution and highlight the potential of hiPSC-derived systems as powerful platforms for modelling human eye development and disease.
Collapse
Affiliation(s)
- Laura Howard
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, Wales, UK
- Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Yuki Ishikawa
- Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomohiko Katayama
- Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Sung-Joon Park
- Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Matthew J Hill
- Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Derek J Blake
- Centre for Neuropsychiatric Genetics and Genomics, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Kohji Nishida
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan.
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan.
| | - Ryuhei Hayashi
- Department of Stem Cells and Applied Medicine, Osaka University Graduate School of Medicine, Osaka, Japan.
- Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Andrew J Quantock
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, Wales, UK
| |
Collapse
|
37
|
Laub V, Nan E, Elias L, Donaldson IJ, Bentsen M, Rusling LA, Schupp J, Lun JH, Plate KH, Looso M, Langer JD, Günther S, Bobola N, Schulte D. Integrated multi-omics analysis of PBX1 in mouse adult neural stem- and progenitor cells identifies a transcriptional module that functionally links PBX1 to TCF3/4. Nucleic Acids Res 2024; 52:12262-12280. [PMID: 39377397 PMCID: PMC11551771 DOI: 10.1093/nar/gkae864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 08/22/2024] [Accepted: 09/23/2024] [Indexed: 10/09/2024] Open
Abstract
Developmental transcription factors act in networks, but how these networks achieve cell- and tissue specificity is still poorly understood. Here, we explored pre-B cell leukemia homeobox 1 (PBX1) in adult neurogenesis combining genomic, transcriptomic, and proteomic approaches. ChIP-seq analysis uncovered PBX1 binding to numerous genomic sites. Integration of PBX1 ChIP-seq with ATAC-seq data predicted interaction partners, which were subsequently validated by mass spectrometry. Whole transcriptome spatial RNA analysis revealed shared expression dynamics of Pbx1 and interacting factors. Among these were class I bHLH proteins TCF3 and TCF4. RNA-seq following Pbx1, Tcf3 or Tcf4 knockdown identified proliferation- and differentiation associated genes as shared targets, while sphere formation assays following knockdown argued for functional cooperativity of PBX1 and TCF3 in progenitor cell proliferation. Notably, while physiological PBX1-TCF interaction has not yet been described, chromosomal translocation resulting in genomic TCF3::PBX1 fusion characterizes a subtype of acute lymphoblastic leukemia. Introducing Pbx1 into Nalm6 cells, a pre-B cell line expressing TCF3 but lacking PBX1, upregulated the leukemogenic genes BLK and NOTCH3, arguing that functional PBX1-TCF cooperativity likely extends to hematopoiesis. Our study hence uncovers a transcriptional module orchestrating the balance between progenitor cell proliferation and differentiation in adult neurogenesis with potential implications for leukemia etiology.
Collapse
Affiliation(s)
- Vera Laub
- Goethe University, University Hospital Frankfurt, Neurological Institute (Edinger Institute), 60528 Frankfurt am Main, Germany
| | - Elisabeth Nan
- Goethe University, University Hospital Frankfurt, Neurological Institute (Edinger Institute), 60528 Frankfurt am Main, Germany
| | - Lena Elias
- Goethe University, University Hospital Frankfurt, Neurological Institute (Edinger Institute), 60528 Frankfurt am Main, Germany
| | - Ian J Donaldson
- University of Manchester, Faculty of Biology, Medicine and Health, Bioinformatics Core Facility, Manchester, M13 9PT, UK
| | - Mette Bentsen
- Max Planck Institute for Heart and Lung Research, Bioinformatics Core Unit (BCU), 61231 Bad Nauheim, Germany
| | - Leona A Rusling
- Max Planck Institute for Biophysics, Proteomics, and Max Planck Institute for Brain Research, 60438 Frankfurt am Main, Germany
| | - Jonathan Schupp
- Goethe University, University Hospital Frankfurt, Neurological Institute (Edinger Institute), 60528 Frankfurt am Main, Germany
- Goethe University, Frankfurt Cancer Institute, 60528 Frankfurt am Main, Germany
| | - Jennifer H Lun
- Goethe University, University Hospital Frankfurt, Neurological Institute (Edinger Institute), 60528 Frankfurt am Main, Germany
- Goethe University, Frankfurt Cancer Institute, 60528 Frankfurt am Main, Germany
| | - Karl H Plate
- Goethe University, University Hospital Frankfurt, Neurological Institute (Edinger Institute), 60528 Frankfurt am Main, Germany
- Goethe University, Frankfurt Cancer Institute, 60528 Frankfurt am Main, Germany
| | - Mario Looso
- Max Planck Institute for Heart and Lung Research, Bioinformatics Core Unit (BCU), 61231 Bad Nauheim, Germany
| | - Julian D Langer
- Max Planck Institute for Biophysics, Proteomics, and Max Planck Institute for Brain Research, 60438 Frankfurt am Main, Germany
| | - Stefan Günther
- Max Planck Institute for Heart and Lung Research, Bioinformatics and Deep Sequencing Platform, 61231 Bad Nauheim, Germany
| | - Nicoletta Bobola
- University of Manchester, Faculty of Biology, Medicine and Health, Manchester, M13 9PT, UK
| | - Dorothea Schulte
- Goethe University, University Hospital Frankfurt, Neurological Institute (Edinger Institute), 60528 Frankfurt am Main, Germany
| |
Collapse
|
38
|
Tommasini D, Yoshimatsu T, Baden T, Shekhar K. Comparative transcriptomic insights into the evolutionary origin of the tetrapod double cone. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621990. [PMID: 39574734 PMCID: PMC11580882 DOI: 10.1101/2024.11.04.621990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2024]
Abstract
The tetrapod double cone is a pair of tightly associated cones called the "principal" and the "accessory" member. It is found in amphibians, reptiles, and birds, as well as monotreme and marsupial mammals but is absent in fish and eutherian mammals. To explore the potential evolutionary origins of the double cone, we analyzed single-cell and -nucleus transcriptomic atlases of photoreceptors from six vertebrate species: zebrafish, chicken, lizard, opossum, ground squirrel, and human. Computational analyses separated the principal and accessory members in chicken and lizard, identifying molecular signatures distinguishing either member from single cones and rods in the same species. Comparative transcriptomic analyses suggest that both the principal and accessory originated from ancestral red cones. Furthermore, the gene expression variation among cone subtypes mirrors their spectral order (red → green → blue → UV), suggesting a constraint in their order of emergence during evolution. Finally, we find that rods are equally dissimilar to all cone types, suggesting that they emerged before the spectral diversification of cones.
Collapse
Affiliation(s)
- Dario Tommasini
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Takeshi Yoshimatsu
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis School of Medicine, St Louis, MO, USA
| | - Tom Baden
- Center for Sensory Neuroscience and Computation, Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Karthik Shekhar
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
- Chemical and Biomolecular Engineering Department, University of California, Berkeley, CA, USA
- Vision Sciences Graduate Program; Center for Computational Biology; Biophysics Graduate Group, University of California, Berkeley, CA, USA
- Biological Systems and Engineering Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| |
Collapse
|
39
|
D'Souza SP, Upton BA, Eldred KC, Glass I, Nayak G, Grover K, Ahmed A, Nguyen MT, Hu YC, Gamlin P, Lang RA. Developmental control of rod number via a light-dependent retrograde pathway from intrinsically photosensitive retinal ganglion cells. Dev Cell 2024; 59:2897-2911.e6. [PMID: 39142280 PMCID: PMC11537824 DOI: 10.1016/j.devcel.2024.07.018] [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: 02/27/2024] [Revised: 06/07/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024]
Abstract
Photoreception is essential for the development of the visual system, shaping vision's first synapse to cortical development. Here, we find that the lighting environment controls developmental rod apoptosis via Opn4-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs). Using genetics, sensory environment manipulations, and computational approaches, we establish a pathway where light-dependent glutamate released from ipRGCs is detected via a transiently expressed glutamate receptor (Grik3) on rod precursors within the inner retina. Communication between these cells is mediated by hybrid neurites on ipRGCs that sense light before eye opening. These structures span the ipRGC-rod precursor distance over development and contain the machinery for photoreception (Opn4) and neurotransmitter release (Vglut2 & Syp). Assessment of the human gestational retina identifies conserved hallmarks of an ipRGC-to-rod axis, including displaced rod precursors, transient GRIK3 expression, and ipRGCs with deep-projecting neurites. This analysis defines an adaptive retrograde pathway linking the sensory environment to rod precursors via ipRGCs prior to eye opening.
Collapse
Affiliation(s)
- Shane P D'Souza
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| | - Brian A Upton
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Kiara C Eldred
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Ian Glass
- Birth Defects Research Laboratory, Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98109, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA 98195, USA
| | - Gowri Nayak
- Transgenic Animal and Genome Editing Core, Department of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Kassidy Grover
- Division of Hematology and Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Neuroscience Graduate Program, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Abdulla Ahmed
- Medical Doctor (M.D.) Training Program, George Washington University School of Medicine, Washington, DC 20052, USA
| | - Minh-Thanh Nguyen
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Yueh-Chiang Hu
- Transgenic Animal and Genome Editing Core, Department of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Paul Gamlin
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Richard A Lang
- Division of Pediatric Ophthalmology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Science of Light Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Abrahamson Pediatric Eye Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Ophthalmology, University of Cincinnati, Cincinnati, OH 45229, USA.
| |
Collapse
|
40
|
Ferrena A, Zhang X, Shrestha R, Zheng D, Liu W. Six3 and Six6 jointly control diverse target genes in multiple cell populations over developmental trajectories of mouse embryonic retinal progenitor cells. PLoS One 2024; 19:e0308839. [PMID: 39446806 PMCID: PMC11500937 DOI: 10.1371/journal.pone.0308839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 08/01/2024] [Indexed: 10/26/2024] Open
Abstract
How tissue-specific progenitor cells generate adult tissues is a puzzle in organogenesis. Using single-cell RNA sequencing of control and Six3 and Six6 compound-mutant mouse embryonic eyecups, we demonstrated that these two closely related transcription factors jointly control diverse target genes in multiple cell populations over the developmental trajectories of mouse embryonic retinal progenitor cells. In the Uniform Manifold Approximation and Projection for Dimension Reduction (UMAP) graph of control retinas, naïve retinal progenitor cells had two major trajectories leading to ciliary margin cells and retinal neurons, respectively. The ciliary margin trajectory was from naïve retinal progenitor cells in the G1 phase directly to ciliary margin cells, whereas the neuronal trajectory went through an intermediate neurogenic state marked by Atoh7 expression. Neurogenic retinal progenitor cells (Atoh7+) were still proliferative; early retinal neurons branched out from Atoh7+ retina progenitor cells in the G1 phase. Upon Six3 and Six6 dual deficiency, both naïve and neurogenic retinal progenitor cells were defective, ciliary margin differentiation was enhanced, and multi-lineage neuronal differentiation was disrupted. An ectopic neuronal trajectory lacking the Atoh7+ state led to ectopic neurons. Additionally, Wnt signaling was upregulated, whereas FGF signaling was downregulated. Notably, Six3 and Six6 proteins occupied the loci of diverse genes that were differentially expressed in distinct cell populations, and expression of these genes was significantly altered upon Six3 and Six6 dual deficiency. Our findings provide deeper insight into the molecular mechanisms underlying early retinal differentiation in mammals.
Collapse
Affiliation(s)
- Alexander Ferrena
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Xusheng Zhang
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Rupendra Shrestha
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, United States of America
- The Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Wei Liu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, United States of America
- The Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
41
|
Bhattacharya S, Yang TS, Nabit BP, Krystofiak ES, Rex TS, Chaum E. Prominin-1 Knockdown Causes RPE Degeneration in a Mouse Model. Cells 2024; 13:1761. [PMID: 39513868 PMCID: PMC11545618 DOI: 10.3390/cells13211761] [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: 08/07/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
There are currently no effective treatments for retinal pigment epithelial (RPE) cell loss in atrophic AMD (aAMD). However, our research on Prominin-1 (Prom1), a known structural protein in photoreceptors (PRs), has revealed its distinct role in RPE and offers promising insights. While pathogenic Prom1 mutations have been linked to macular diseases with RPE atrophy, the broader physiological impact of dysfunctional Prom1 in RPE loss is unclear. We have shown that Prom1 plays a crucial role in regulating autophagy and cellular homeostasis in human and mouse RPE (mRPE) cells in vitro. Nevertheless, a comprehensive understanding of its in vivo expression and function in mRPE remains to be elucidated. To characterize Prom1 expression in RPE in situ, we used RNAscope assays and immunogold electron microscopy (EM). Our use of chromogenic and fluorescent RNAscope assays in albino and C57BL/6J mouse retinal sections has revealed Prom1 mRNA expression in perinuclear regions in mRPE in situ. Immunogold EM imaging showed Prom1 expression in RPE cytoplasm and mitochondria. To confirm Prom1 expression in RPE, we interrogated human RPE single-cell RNA-sequencing datasets using an online resource, Spectacle. Our analysis showed Prom1 expression in human RPE. To investigate Prom1's function in RPE homeostasis, we performed RPE-specific Prom1 knockdown (KD) using subretinal injections of AAV2/1.CMV.saCas9.U6.Prom1gRNA in male and female mice. Our data show that RPE-specific Prom1-KD in vivo resulted in abnormal RPE morphology, subretinal fluid accumulation, and secondary PR loss. These changes were associated with patchy RPE cell death and reduced a-wave amplitude, indicating retinal degeneration. Our findings underscore the central role of Prom1 in cell-autonomous mRPE homeostasis. The implications of Prom1-KD causing aAMD-like RPE defects and retinal degeneration in a mouse model are significant and could lead to novel treatments for aAMD.
Collapse
Grants
- S10 OD034315 NIH HHS
- P30CA068485, S10OD023475-01A1, S10 OD016355, IS1BX003154, CA68485, DK20593, DK58404, DK59637, EY08126, and 1S100D034315-01. International Retina Research Foundation, the Potocsnak family gift to the Vanderbilt Eye Institute, the Margy Ann and J Donald M Gass Chair endowment, and an unrestricted departmental research grant from Research to Prevent Blindness, Inc (New York, NY).
Collapse
Affiliation(s)
- Sujoy Bhattacharya
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA (E.C.)
| | - Tzushan Sharon Yang
- Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Bretton P. Nabit
- Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Evan S. Krystofiak
- Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, USA
| | - Tonia S. Rex
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA (E.C.)
| | - Edward Chaum
- Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37232, USA (E.C.)
- Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
42
|
Scheri KC, Tedeschi T, Fawzi AA. Single Cell Isolation from Human Diabetic Fibrovascular Membranes for Single-Cell RNA Sequencing. Bio Protoc 2024; 14:e5096. [PMID: 39512888 PMCID: PMC11540046 DOI: 10.21769/bioprotoc.5096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 11/15/2024] Open
Abstract
Single-cell transcriptomic analyses have emerged as very powerful tools to query the gene expression changes at the single-cell level in physiological and pathological conditions. The quality of the analysis is heavily dependent on tissue digestion protocols, with the goal of preserving thousands of single live cells to submit to the subsequent processing steps and analysis. Multiple digestion protocols that use different enzymes to digest the tissues have been described. Harsh digestion can damage certain cell types, but this might be required to digest especially fibrotic tissue as in our experimental condition. In this paper, we summarize a collagenase type I digestion protocol for preparing the single-cell suspension from fibrovascular tissues surgically removed from patients with proliferative diabetic retinopathy (PDR) for single-cell RNA sequencing (scRNA-Seq) analyses. We also provide a detailed description of the data analysis that we implemented in a previously published study. Key features • Single-cell suspension from fibrovascular membranes isolated from PDR patients. • Single-cell RNA sequencing analyses performed using Seurat package in RStudio. • Trajectory analyses or pseudotime analyses to study the trajectory over (pseudo)time of specific cell types. • This protocol requires Illumina HiSEQ4000 instrument and knowledge of R and RStudio language for the analyses.
Collapse
Affiliation(s)
- Katia Corano Scheri
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Thomas Tedeschi
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Amani A Fawzi
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
43
|
Scheepers R, Levi NL, Araujo RP. A distributed integral control mechanism for regulation of cholesterol concentration in the human retina. ROYAL SOCIETY OPEN SCIENCE 2024; 11:240432. [PMID: 39479233 PMCID: PMC11521609 DOI: 10.1098/rsos.240432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/24/2024] [Accepted: 08/11/2024] [Indexed: 11/02/2024]
Abstract
Tight homeostatic control of cholesterol concentration within the complex tissue microenvironment of the retina is the hallmark of a healthy eye. By contrast, dysregulation of biochemical mechanisms governing retinal cholesterol homeostasis likely contributes to the aetiology and progression of age-related macular degeneration (AMD). While the signalling mechanisms maintaining cellular cholesterol homeostasis are well-studied, a systems-level description of molecular interactions regulating cholesterol balance within the human retina remains elusive. Here, we provide a comprehensive overview of all currently-known molecular-level interactions involved in cholesterol regulation across the major compartments of the human retina, encompassing the retinal pigment epithelium (RPE), photoreceptor cell layer, Müller cell layer and Bruch's membrane. We develop a comprehensive chemical reaction network (CRN) of these interactions, involving 71 molecular species, partitioned into 10 independent subnetworks. These subnetworks collectively ensure robust homeostasis of 14 forms of cholesterol across distinct retinal cellular compartments. We provide mathematical evidence that three independent antithetic integral feedback controllers tightly regulate ER cholesterol in retinal cells, with additional independent mechanisms extending this regulation to other forms of cholesterol throughout the retina. Our novel mathematical model of retinal cholesterol regulation provides a framework for understanding the mechanisms of cholesterol dysregulation in diseased eyes and for exploring potential therapeutic strategies.
Collapse
Affiliation(s)
- Ronél Scheepers
- School of Mathematical Sciences, Queensland University of Technology (QUT), Brisbane4000, Australia
| | - Noa L. Levi
- School of Mathematics and Statistics, University of Melbourne, Victoria3010, Australia
| | - Robyn P. Araujo
- School of Mathematics and Statistics, University of Melbourne, Victoria3010, Australia
| |
Collapse
|
44
|
Blackshaw S, Qian J, Hyde DR. New pathways to neurogenesis: Insights from injury-induced retinal regeneration. Bioessays 2024; 46:e2400133. [PMID: 38990084 PMCID: PMC11897919 DOI: 10.1002/bies.202400133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024]
Abstract
The vertebrate retina is a tractable system for studying control of cell neurogenesis and cell fate specification. During embryonic development, retinal neurogenesis is under strict temporal regulation, with cell types generated in fixed but overlapping temporal intervals. The temporal sequence and relative numbers of retinal cell types generated during development are robust and show minimal experience-dependent variation. In many cold-blooded vertebrates, acute retinal injury induces a different form of neurogenesis, where Müller glia reprogram into retinal progenitor-like cells that selectively regenerate retinal neurons lost to injury. The extent to which the molecular mechanisms controlling developmental and injury-induced neurogenesis resemble one another has long been unclear. However, a recent study in zebrafish has shed new light on this question, using single-cell multiomic analysis to show that selective loss of different retinal cell types induces the formation of fate-restricted Müller glia-derived progenitors that differ both from one another and from progenitors in developing retina. Here, we discuss the broader implications of these findings, and their possible therapeutic relevance.
Collapse
Affiliation(s)
- Seth Blackshaw
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jiang Qian
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David R. Hyde
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
- Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, USA
- Center for Zebrafish Research, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
45
|
Zeng X, Gyoja F, Cui Y, Loza M, Kusakabe T, Nakai K. Comparative single-cell transcriptomic analysis reveals putative differentiation drivers and potential origin of vertebrate retina. NAR Genom Bioinform 2024; 6:lqae149. [PMID: 39534499 PMCID: PMC11555436 DOI: 10.1093/nargab/lqae149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 10/10/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Despite known single-cell expression profiles in vertebrate retinas, understanding of their developmental and evolutionary expression patterns among homologous cell classes remains limited. We examined and compared approximately 240 000 retinal cells from four species and found significant similarities among homologous cell classes, indicating inherent regulatory patterns. To understand these shared patterns, we constructed gene regulatory networks for each developmental stage for three of these species. We identified 690 regulons governed by 530 regulators across three species, along with 10 common cell class-specific regulators and 16 highly preserved regulons. RNA velocity analysis pinpointed conserved putative driver genes and regulators to retinal cell differentiation in both mouse and zebrafish. Investigation of the origins of retinal cells by examining conserved expression patterns between vertebrate retinal cells and invertebrate Ciona intestinalis photoreceptor-related cells implied functional similarities in light transduction mechanisms. Our findings offer insights into the evolutionarily conserved regulatory frameworks and differentiation drivers of vertebrate retinal cells.
Collapse
Affiliation(s)
- Xin Zeng
- Department of Computational Biology and Medical Sciences, The University of Tokyo, Kashiwa 277-8563, Japan
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Fuki Gyoja
- Institute for Integrative Neurobiology and Department of Biology, Konan University, Kobe 658-8501, Japan
| | - Yang Cui
- Department of Computational Biology and Medical Sciences, The University of Tokyo, Kashiwa 277-8563, Japan
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Martin Loza
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Takehiro G Kusakabe
- Institute for Integrative Neurobiology and Department of Biology, Konan University, Kobe 658-8501, Japan
| | - Kenta Nakai
- Department of Computational Biology and Medical Sciences, The University of Tokyo, Kashiwa 277-8563, Japan
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
46
|
Shayler DW, Stachelek K, Cambier L, Lee S, Bai J, Reid MW, Weisenberger DJ, Bhat B, Aparicio JG, Kim Y, Singh M, Bay M, Thornton ME, Doyle EK, Fouladian Z, Erberich SG, Grubbs BH, Bonaguidi MA, Craft CM, Singh HP, Cobrinik D. Identification and characterization of early human photoreceptor states and cell-state-specific retinoblastoma-related features. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.28.530247. [PMID: 38915659 PMCID: PMC11195049 DOI: 10.1101/2023.02.28.530247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Human cone photoreceptors differ from rods and serve as the retinoblastoma cell-of-origin, yet the developmental basis for their distinct behaviors is poorly understood. Here, we used deep full-length single-cell RNA-sequencing to distinguish post-mitotic cone and rod developmental states and identify cone-specific features that contribute to retinoblastomagenesis. The analyses revealed early post-mitotic cone- and rod-directed populations characterized by higher THRB or NRL regulon activities, an immature photoreceptor precursor population with concurrent cone and rod gene and regulon expression, and distinct early and late cone and rod maturation states distinguished by maturation-associated declines in RAX regulon activity. Unexpectedly, both L/M cone and rod precursors co-expressed NRL and THRB RNAs, yet they differentially expressed functionally antagonistic NRL and THRB isoforms and prematurely terminated THRB transcripts. Early L/M cone precursors exhibited successive expression of several lncRNAs along with MYCN, which composed the seventh most L/M-cone-specific regulon, and SYK, which contributed to the early cone precursors' proliferative response to RB1 loss. These findings reveal previously unrecognized photoreceptor precursor states and a role for early cone-precursor-intrinsic SYK expression in retinoblastoma initiation.
Collapse
Affiliation(s)
- Dominic W.H. Shayler
- The Vision Center, Department of Surgery, and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Development, Stem Cell, and Regenerative Medicine Program, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kevin Stachelek
- The Vision Center, Department of Surgery, and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Cancer Biology and Genomics Program, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Linda Cambier
- The Vision Center, Department of Surgery, and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Sunhye Lee
- The Vision Center, Department of Surgery, and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Jinlun Bai
- The Vision Center, Department of Surgery, and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Development, Stem Cell, and Regenerative Medicine Program, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mark W. Reid
- The Vision Center, Department of Surgery, and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Daniel J. Weisenberger
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Bhavana Bhat
- The Vision Center, Department of Surgery, and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Jennifer G. Aparicio
- The Vision Center, Department of Surgery, and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Yeha Kim
- The Vision Center, Department of Surgery, and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Mitali Singh
- The Vision Center, Department of Surgery, and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Maxwell Bay
- Development, Stem Cell, and Regenerative Medicine Program, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Matthew E. Thornton
- Maternal-Fetal Medicine Division of the Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Eamon K. Doyle
- Department of Radiology and The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Zachary Fouladian
- The Vision Center, Department of Surgery, and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Development, Stem Cell, and Regenerative Medicine Program, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Stephan G. Erberich
- Department of Radiology and The Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brendan H. Grubbs
- Maternal-Fetal Medicine Division of the Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Michael A. Bonaguidi
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Development, Stem Cell, and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Cheryl Mae Craft
- Department of Integrative Anatomical Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Hardeep P. Singh
- The Vision Center, Department of Surgery, and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - David Cobrinik
- The Vision Center, Department of Surgery, and Saban Research Institute, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- USC Roski Eye Institute, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
47
|
Hack SJ, Petereit J, Tseng KAS. Temporal Transcriptomic Profiling of the Developing Xenopus laevis Eye. Cells 2024; 13:1390. [PMID: 39195278 PMCID: PMC11352439 DOI: 10.3390/cells13161390] [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: 07/10/2024] [Revised: 08/06/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024] Open
Abstract
Retinal progenitor cells (RPCs) are a multipotent and highly proliferative population that give rise to all retinal cell types during organogenesis. Defining their molecular signature is a key step towards identifying suitable approaches to treat visual impairments. Here, we performed RNA sequencing of whole eyes from Xenopus at three embryonic stages and used differential expression analysis to define the transcriptomic profiles of optic tissues containing proliferating and differentiating RPCs during retinogenesis. Gene Ontology and KEGG pathway analyses showed that genes associated with developmental pathways (including Wnt and Hedgehog signaling) were upregulated during the period of active RPC proliferation in early retinal development (Nieuwkoop Faber st. 24 and 27). Developing eyes had dynamic expression profiles and shifted to enrichment for metabolic processes and phototransduction during RPC progeny specification and differentiation (st. 35). Furthermore, conserved adult eye regeneration genes were also expressed during early retinal development, including sox2, pax6, nrl, and Notch signaling components. The eye transcriptomic profiles presented here span RPC proliferation to retinogenesis and include regrowth-competent stages. Thus, our dataset provides a rich resource to uncover molecular regulators of RPC activity and will allow future studies to address regulators of RPC proliferation during eye repair and regrowth.
Collapse
Affiliation(s)
- Samantha J. Hack
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA
| | - Juli Petereit
- Nevada Bioinformatics Center, University of Nevada, Reno, NV 89557, USA
| | | |
Collapse
|
48
|
Zuo Z, Cheng X, Ferdous S, Shao J, Li J, Bao Y, Li J, Lu J, Jacobo Lopez A, Wohlschlegel J, Prieve A, Thomas MG, Reh TA, Li Y, Moshiri A, Chen R. Single cell dual-omic atlas of the human developing retina. Nat Commun 2024; 15:6792. [PMID: 39117640 PMCID: PMC11310509 DOI: 10.1038/s41467-024-50853-5] [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: 10/06/2023] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
The development of the retina is under tight temporal and spatial control. To gain insights into the molecular basis of this process, we generate a single-nuclei dual-omic atlas of the human developing retina with approximately 220,000 nuclei from 14 human embryos and fetuses aged between 8 and 23-weeks post-conception with matched macular and peripheral tissues. This atlas captures all major cell classes in the retina, along with a large proportion of progenitors and cell-type-specific precursors. Cell trajectory analysis reveals a transition from continuous progression in early progenitors to a hierarchical development during the later stages of cell type specification. Both known and unrecorded candidate transcription factors, along with gene regulatory networks that drive the transitions of various cell fates, are identified. Comparisons between the macular and peripheral retinae indicate a largely consistent yet distinct developmental pattern. This atlas offers unparalleled resolution into the transcriptional and chromatin accessibility landscapes during development, providing an invaluable resource for deeper insights into retinal development and associated diseases.
Collapse
Affiliation(s)
- Zhen Zuo
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Xuesen Cheng
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Salma Ferdous
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Jianming Shao
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Jin Li
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Yourong Bao
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Jean Li
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Jiaxiong Lu
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Antonio Jacobo Lopez
- Department of Ophthalmology & Vision Science, UC Davis School of Medicine, 4860 Y St, Sacramento, CA, USA
| | - Juliette Wohlschlegel
- Department of Biological Structure, University of Washington, 1410 NE Campus Pkwy, Seattle, WA, USA
| | - Aric Prieve
- Department of Biological Structure, University of Washington, 1410 NE Campus Pkwy, Seattle, WA, USA
| | - Mervyn G Thomas
- Ulverscroft Eye Unit, School of Psychology and Vision Sciences, The University of Leicester, Leicester, UK
| | - Thomas A Reh
- Department of Biological Structure, University of Washington, 1410 NE Campus Pkwy, Seattle, WA, USA
| | - Yumei Li
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Ala Moshiri
- Department of Ophthalmology & Vision Science, UC Davis School of Medicine, 4860 Y St, Sacramento, CA, USA
| | - Rui Chen
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA.
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA.
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA.
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California Irvine School of Medicine, Irvine, USA.
| |
Collapse
|
49
|
Luo S, Alwattar B, Li Q, Bora K, Blomfield AK, Lin J, Fulton A, Chen J, Agrawal PB. HBS1L deficiency causes retinal dystrophy in a child and in a mouse model associated with defective development of photoreceptor cells. Dis Model Mech 2024; 17:dmm050557. [PMID: 38966981 PMCID: PMC11317091 DOI: 10.1242/dmm.050557] [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: 10/19/2023] [Accepted: 06/21/2024] [Indexed: 07/06/2024] Open
Abstract
Inherited retinal diseases encompass a genetically diverse group of conditions caused by variants in genes critical to retinal function, including handful of ribosome-associated genes. This study focuses on the HBS1L gene, which encodes for the HBS1-like translational GTPase that is crucial for ribosomal rescue. We have reported a female child carrying biallelic HBS1L variants, manifesting with poor growth and neurodevelopmental delay. Here, we describe the ophthalmologic findings in the patient and in Hbs1ltm1a/tm1a hypomorph mice and describe the associated microscopic and molecular perturbations. The patient has impaired visual function, showing dampened amplitudes of a- and b-waves in both rod- and cone-mediated responses. Hbs1ltm1a/tm1a mice exhibited profound thinning of the entire retina, specifically of the outer photoreceptor layer, due to extensive photoreceptor cell apoptosis. Loss of Hbs1l resulted in comprehensive proteomic alterations by mass spectrometry analysis, with an increase in the levels of 169 proteins and a decrease in the levels of 480 proteins, including rhodopsin (Rho) and peripherin 2 (Prph2). Gene Ontology biological process and gene set enrichment analyses reveal that the downregulated proteins are primarily involved in phototransduction, cilium assembly and photoreceptor cell development. These findings underscore the importance of ribosomal rescue proteins in maintaining retinal health, particularly in photoreceptor cells.
Collapse
Affiliation(s)
- Shiyu Luo
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL 33136, USA
- Division of Genetics and Genomics and The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Bilal Alwattar
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Qifei Li
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL 33136, USA
- Division of Genetics and Genomics and The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kiran Bora
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Alexandra K. Blomfield
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jasmine Lin
- Division of Genetics and Genomics and The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Anne Fulton
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jing Chen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Pankaj B. Agrawal
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Holtz Children's Hospital, Jackson Health System, Miami, FL 33136, USA
- Division of Genetics and Genomics and The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
50
|
Bergmans S, Noel NCL, Masin L, Harding EG, Krzywańska AM, De Schutter JD, Ayana R, Hu C, Arckens L, Ruzycki PA, MacDonald RB, Clark BS, Moons L. Age-related dysregulation of the retinal transcriptome in African turquoise killifish. Aging Cell 2024; 23:e14192. [PMID: 38742929 PMCID: PMC11320354 DOI: 10.1111/acel.14192] [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: 03/06/2024] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 05/16/2024] Open
Abstract
Age-related vision loss caused by retinal neurodegenerative pathologies is becoming more prevalent in our ageing society. To understand the physiological and molecular impact of ageing on retinal homeostasis, we used the short-lived African turquoise killifish, a model known to naturally develop central nervous system (CNS) ageing hallmarks and vision loss. Bulk and single-cell RNA-sequencing (scRNAseq) of three age groups (6-, 12-, and 18-week-old) identified transcriptional ageing fingerprints in the killifish retina, unveiling pathways also identified in the aged brain, including oxidative stress, gliosis, and inflammageing. These findings were comparable to observations in the ageing mouse retina. Additionally, transcriptional changes in genes related to retinal diseases, such as glaucoma and age-related macular degeneration, were observed. The cellular heterogeneity in the killifish retina was characterized, confirming the presence of all typical vertebrate retinal cell types. Data integration from age-matched samples between the bulk and scRNAseq experiments revealed a loss of cellular specificity in gene expression upon ageing, suggesting potential disruption in transcriptional homeostasis. Differential expression analysis within the identified cell types highlighted the role of glial/immune cells as important stress regulators during ageing. Our work emphasizes the value of the fast-ageing killifish in elucidating molecular signatures in age-associated retinal disease and vision decline. This study contributes to the understanding of how age-related changes in molecular pathways may impact CNS health, providing insights that may inform future therapeutic strategies for age-related pathologies.
Collapse
Affiliation(s)
- Steven Bergmans
- Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research GroupKU Leuven, Leuven Brain InstituteLeuvenBelgium
| | | | - Luca Masin
- Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research GroupKU Leuven, Leuven Brain InstituteLeuvenBelgium
| | - Ellen G. Harding
- John F Hardesty, MD Department of Ophthalmology and Visual SciencesWashington University School of MedicineSaint LouisMissouriUSA
| | | | - Julie D. De Schutter
- Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research GroupKU Leuven, Leuven Brain InstituteLeuvenBelgium
| | - Rajagopal Ayana
- Department of Biology, Animal Physiology and Neurobiology Section, Laboratory of Neuroplasticity and NeuroproteomicsKU Leuven, Leuven Brain InstituteLeuvenBelgium
| | - Chi‐Kuo Hu
- Department of Biochemistry and Cell BiologyStony Brook UniversityStony BrookUSA
| | - Lut Arckens
- Department of Biology, Animal Physiology and Neurobiology Section, Laboratory of Neuroplasticity and NeuroproteomicsKU Leuven, Leuven Brain InstituteLeuvenBelgium
| | - Philip A. Ruzycki
- John F Hardesty, MD Department of Ophthalmology and Visual SciencesWashington University School of MedicineSaint LouisMissouriUSA
- Department of GeneticsWashington University School of MedicineSaint LouisMissouriUSA
| | | | - Brian S. Clark
- John F Hardesty, MD Department of Ophthalmology and Visual SciencesWashington University School of MedicineSaint LouisMissouriUSA
- Department of Developmental BiologyWashington University School of MedicineSaint LouisMissouriUSA
- Center of Regenerative MedicineCenter of Regenerative Medicine, Washington University School of MedicineSaint LouisMissouriUSA
| | - Lieve Moons
- Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research GroupKU Leuven, Leuven Brain InstituteLeuvenBelgium
| |
Collapse
|