1
|
Noero J, Weber M, Chassaing N, Gaston V, Plaisancié J, Chesneau B. Genetic screening of the RNA-binding protein RBM24 and its binding sites in the SOX2 3' untranslated region in a cohort of 50 patients with micro-anophthalmia. Ophthalmic Genet 2025; 46:256-260. [PMID: 39957481 DOI: 10.1080/13816810.2025.2467334] [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: 06/27/2024] [Revised: 01/27/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
Microphthalmia and anophthalmia (M/A) are rare congenital eye anomalies with a birth prevalence of up to 1 in 10,000 births. The etiology of M/A can involve environmental and/or genetic factors, with a genetic origin identified in approximately 50% of cases through analysis of key genes. The transcription factor SOX2 is the most commonly implicated gene, accounting for around 15% of M/A cases. Recent studies have shown that the RNA-binding protein Rbm24 post-transcriptionally regulates Sox2 expression in mice and zebrafish, with Rbm24 null models exhibiting eye phenotypes in both species. Rbm24 can bind to Sox2 mRNA via three AU-Rich elements (AREs) located in its 3' untranslated region (UTR). In this study, we aimed to determine whether pathogenic variants within RBM24 or the SOX2 3'UTR AREs were present in a cohort of 50 individuals with M/A with no identified genetic cause for their condition. Despite the ocular defects observed in animal models, we did not detect any variant of interest in these candidate regions in our cohort. Although we cannot exclude the involvement of pathogenic variations in RBM24 or the SOX2 3'UTR AREs in ocular developmental defects, our study shows that they are unlikely to represent a frequent cause of M/A.
Collapse
Affiliation(s)
- Julien Noero
- Laboratoire de Référence (LBMR) des anomalies malformatives de l'œil, Institut Fédératif de Biologie (IFB), CHU de Toulouse, Toulouse, France
- CNRS UMR5077, Centre de Biologie Intégrative, Université Toulouse III, Toulouse, France
| | - Mathys Weber
- Laboratoire de Référence (LBMR) des anomalies malformatives de l'œil, Institut Fédératif de Biologie (IFB), CHU de Toulouse, Toulouse, France
| | - Nicolas Chassaing
- Laboratoire de Référence (LBMR) des anomalies malformatives de l'œil, Institut Fédératif de Biologie (IFB), CHU de Toulouse, Toulouse, France
- Centre de Référence des Affections Rares en Génétique Ophtalmologique, CARGO, CHU de Toulouse, Toulouse, France
| | - Véronique Gaston
- Laboratoire de Référence (LBMR) des anomalies malformatives de l'œil, Institut Fédératif de Biologie (IFB), CHU de Toulouse, Toulouse, France
| | - Julie Plaisancié
- Laboratoire de Référence (LBMR) des anomalies malformatives de l'œil, Institut Fédératif de Biologie (IFB), CHU de Toulouse, Toulouse, France
- CNRS UMR5077, Centre de Biologie Intégrative, Université Toulouse III, Toulouse, France
- Centre de Référence des Affections Rares en Génétique Ophtalmologique, CARGO, CHU de Toulouse, Toulouse, France
| | - Bertrand Chesneau
- Laboratoire de Référence (LBMR) des anomalies malformatives de l'œil, Institut Fédératif de Biologie (IFB), CHU de Toulouse, Toulouse, France
- CNRS UMR5077, Centre de Biologie Intégrative, Université Toulouse III, Toulouse, France
- Centre de Référence des Affections Rares en Génétique Ophtalmologique, CARGO, CHU de Toulouse, Toulouse, France
| |
Collapse
|
2
|
Atalay K, Kocak I, Sayin N, Cirak B. Corneal endothelial and retinal microvascular changes in pseudoexfoliation syndrome: insights from specular microscopy and OCT angiography. BMC Ophthalmol 2025; 25:202. [PMID: 40217452 PMCID: PMC11992760 DOI: 10.1186/s12886-025-04033-8] [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: 01/16/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
PURPOSE To investigate the correlation between corneal specular microscopy (SM) and macular optical coherence tomography angiography (OCTA) data in patients with pseudoexfoliation syndrome (PXS). METHODS This was a cross-sectional, prospective study. Consecutive patients aged > 45 years with PXS or normal examination results were included. We examined cell density (CD), average cell area (AVG), coefficient of variation (CV), and hexagonality values from the SM. We collected 6 × 6 mm macular OCTA data with a quality score of 6/10 or higher. RESULTS Thirty-five eyes with PXS and 32 healthy eyes were evaluated. The CD, AVG, CV, and hexagonality for the PXS group were 2388.5 ± 368.8 cell/mm2, 435.0 ± 117.6 µm2, 28.9 ± 4.8%, and 67.6 ± 4.8%, respectively. The control group had the following values for CD, AVG, CV, and hexagonality: 2654.5 ± 269.8 cell/mm2, 380.4 ± 38.8 µm2, 29.8 ± 5.1%, and 67.1 ± 4.9%, respectively. No correlation was observed between OCTA and SM findings in the PXS group. We found a slight but significant link between CD and OCTA outer and total field estimations in the control group (p = 0.03 r = -0.25 and p = 0.02, r = 0.27, respectively). AVG and OCTA data in the control group indicated an association in the outer- and full-field analyses (p = 0.03 r = 0.25, p = 0.02, r = 0.27, respectively). CONCLUSION Highlighting the observed lack of correlation between SM and macular OCTA findings in PXS patients in this study emphasizes the necessity of prioritizing early detection of anterior segment changes. The results indicate that immediate monitoring and intervention could reduce undesired consequences and maintain visual function in those affected.
Collapse
Affiliation(s)
- Kursat Atalay
- Kanuni Sultan Suleyman Training and Research Hospital, Eye Clinic, Istanbul, 34303, Atakent Mah. Turgut Ozal Blv. Kucukkcekmece, Turkey.
| | - Ibrahim Kocak
- Kanuni Sultan Suleyman Training and Research Hospital, Eye Clinic, Istanbul, 34303, Atakent Mah. Turgut Ozal Blv. Kucukkcekmece, Turkey
| | - Nihat Sayin
- Kanuni Sultan Suleyman Training and Research Hospital, Eye Clinic, Istanbul, 34303, Atakent Mah. Turgut Ozal Blv. Kucukkcekmece, Turkey
| | - Busra Cirak
- Kanuni Sultan Suleyman Training and Research Hospital, Eye Clinic, Istanbul, 34303, Atakent Mah. Turgut Ozal Blv. Kucukkcekmece, Turkey
| |
Collapse
|
3
|
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
|
4
|
Zheng Y, Stormo GD, Chen S. Aberrant homeodomain-DNA cooperative dimerization underlies distinct developmental defects in two dominant CRX retinopathy models. Genome Res 2025; 35:242-256. [PMID: 39715683 PMCID: PMC11874979 DOI: 10.1101/gr.279340.124] [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: 03/15/2024] [Accepted: 12/10/2024] [Indexed: 12/25/2024]
Abstract
Paired-class homeodomain (HD) transcription factors (TFs) play essential roles in vertebrate development, and their mutations are linked to human diseases. One unique feature of a paired-class HD is cooperative dimerization on specific palindrome DNA sequences. Yet, the functional significance of HD cooperative dimerization in animal development and its dysregulation in diseases remains elusive. Using the retinal TF cone-rod homeobox (CRX) as a model, we have studied how blindness-causing mutations in the paired HD, p.E80A and p.K88N, alter CRX's cooperative dimerization, leading to gene misexpression and photoreceptor developmental deficits in dominant manners. CRXE80A maintains binding at monomeric WT CRX motifs but is deficient in cooperative binding at dimeric motifs. CRXE80A's cooperativity defect impacts the exponential increase of photoreceptor gene expression in terminal differentiation and produces immature, nonfunctional photoreceptors in the Crx E80A retinas. CRXK88N is highly cooperative and localizes to ectopic genomic sites with strong enrichment of dimeric HD motifs. CRXK88N's altered biochemical properties disrupt CRX's ability to direct dynamic chromatin remodeling during development to activate photoreceptor differentiation programs and silence progenitor programs. Our study provides in vitro and in vivo molecular evidence that paired-class HD cooperative dimerization regulates neuronal development and that dysregulation of cooperative binding contributes to severe dominant blinding retinopathies.
Collapse
Affiliation(s)
- Yiqiao Zheng
- Molecular Genetics and Genomics Graduate Program, Division of Biology & Biomedical Sciences, Washington University in St. Louis, St. Louis, Missouri 63110, USA
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis, Saint Louis, Missouri 63110, USA
| | - Gary D Stormo
- Department of Genetics, Washington University in St Louis, Saint Louis, Missouri 63110, USA;
| | - Shiming Chen
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis, Saint Louis, Missouri 63110, USA;
- Department of Developmental Biology, Washington University in St Louis, Saint Louis, Missouri 63110, USA
| |
Collapse
|
5
|
Longakit AN, Bourget H, Van Raamsdonk CD. Mitf over-expression leads to microphthalmia and coloboma in Mitf-cre mice. Exp Eye Res 2025; 251:110209. [PMID: 39694408 DOI: 10.1016/j.exer.2024.110209] [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: 08/21/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
The Mitf transcription factor is a critical regulator of the melanocyte lineage and eye development. Mitf activity in different cell types is controlled in part by ten alternative promoters and their resulting isoforms. A useful tool for melanocyte-based research, Mitf-cre was designed to express Cre from the Mitf-M promoter, which is melanocyte specific. However, Mitf-cre mice are also microphthalmic, perhaps because of insertional mutagenesis or disrupted gene expression. Here, we investigated these possibilities and described the eye phenotype. Targeted locus amplification indicated that the transgene integrated on chromosome 2, in between Spred1 and Meis2. The BAC transgene used to make Mitf-cre was larger than expected, carrying three upstream alternative promoters, Mitf-H, Mitf-D, and Mitf-B, which could express their isoforms intact off the transgene. RT-qPCR using eye tissue demonstrated a 5-fold increase in Mitf transcripts containing exon 1B1b, which is shared by Mitf-H, Mitf-D, and Mitf-B, while Spred1 and Meis2 did not differ in their expression. These findings clarify and support the usage of Mitf-cre in conditional mutagenesis in melanocytes. The specific over-expression of these isoforms, which are preferentially expressed in the RPE, presents a unique resource for those interested in eye development and coloboma.
Collapse
Affiliation(s)
- Anne Nathalie Longakit
- Department of Medical Genetics, University of British Columbia, Life Sciences Institute, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Hannah Bourget
- Department of Medical Genetics, University of British Columbia, Life Sciences Institute, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Catherine D Van Raamsdonk
- Department of Medical Genetics, University of British Columbia, Life Sciences Institute, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
6
|
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
|
7
|
More S, Mallick S, P SS, Bose B. Pax6 expressing neuroectodermal and ocular stem cells: Its role from a developmental biology perspective. Cell Biol Int 2024; 48:1802-1815. [PMID: 39308152 DOI: 10.1002/cbin.12246] [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: 05/17/2024] [Revised: 07/29/2024] [Accepted: 08/16/2024] [Indexed: 11/15/2024]
Abstract
Pax-6 emerges as a critical transcription factor that guides the fate of stem cells towards neural lineages. Its expression influences the differentiation of neural progenitors into diverse neuronal subtypes, glial cells, and other neural cell types. Pax-6 operates with other regulatory factors to ensure the precise patterning and organization of the developing nervous system. The intricate interplay between Pax-6 and other signaling pathways, transcription factors, and epigenetic modifiers underpins the complicated balance between stem cell maintenance, proliferation, and differentiation in neuroectodermal and ocular contexts. Dysfunction of Pax-6 can lead to a spectrum of developmental anomalies, underscoring its importance in these processes. This review highlights the essential role of Pax-6 expression in neuroectodermal and ocular stem cells, shedding light on its significance in orchestrating the intricate journey from stem cell fate determination to the emergence of diverse neural and ocular cell types. The comprehensive understanding of Pax-6 function gained from a developmental biology perspective offers valuable insights into normal development and potential therapeutic avenues for neuroectodermal and ocular disorders.
Collapse
Affiliation(s)
- Shubhangi More
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Sumit Mallick
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Sudheer Shenoy P
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka, India
| |
Collapse
|
8
|
Boudriot E, Gabriel V, Popovic D, Pingen P, Yakimov V, Papiol S, Roell L, Hasanaj G, Xu S, Moussiopoulou J, Priglinger S, Kern C, Schulte EC, Hasan A, Pogarell O, Falkai P, Schmitt A, Schworm B, Wagner E, Keeser D, Raabe FJ. Signature of Altered Retinal Microstructures and Electrophysiology in Schizophrenia Spectrum Disorders Is Associated With Disease Severity and Polygenic Risk. Biol Psychiatry 2024; 96:792-803. [PMID: 38679358 DOI: 10.1016/j.biopsych.2024.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 03/30/2024] [Accepted: 04/14/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Optical coherence tomography and electroretinography studies have revealed structural and functional retinal alterations in individuals with schizophrenia spectrum disorders (SSDs). However, it remains unclear which specific retinal layers are affected; how the retina, brain, and clinical symptomatology are connected; and how alterations of the visual system are related to genetic disease risk. METHODS Optical coherence tomography, electroretinography, and brain magnetic resonance imaging were applied to comprehensively investigate the visual system in a cohort of 103 patients with SSDs and 130 healthy control individuals. The sparse partial least squares algorithm was used to identify multivariate associations between clinical disease phenotype and biological alterations of the visual system. The association of the revealed patterns with individual polygenic disease risk for schizophrenia was explored in a post hoc analysis. In addition, covariate-adjusted case-control comparisons were performed for each individual optical coherence tomography and electroretinography parameter. RESULTS The sparse partial least squares analysis yielded a phenotype-eye-brain signature of SSDs in which greater disease severity, longer duration of illness, and impaired cognition were associated with electrophysiological alterations and microstructural thinning of most retinal layers. Higher individual loading onto this disease-relevant signature of the visual system was significantly associated with elevated polygenic risk for schizophrenia. In case-control comparisons, patients with SSDs had lower macular thickness, thinner retinal nerve fiber and inner plexiform layers, less negative a-wave amplitude, and lower b-wave amplitude. CONCLUSIONS This study demonstrates multimodal microstructural and electrophysiological retinal alterations in individuals with SSDs that are associated with disease severity and individual polygenic burden.
Collapse
Affiliation(s)
- Emanuel Boudriot
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany; Max Planck Institute of Psychiatry, Munich, Germany
| | - Vanessa Gabriel
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - David Popovic
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany; Max Planck Institute of Psychiatry, Munich, Germany
| | - Pauline Pingen
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Vladislav Yakimov
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany; International Max Planck Research School for Translational Psychiatry, Munich, Germany
| | - Sergi Papiol
- Max Planck Institute of Psychiatry, Munich, Germany; Institute of Psychiatric Phenomics and Genomics, LMU Munich, Munich, Germany
| | - Lukas Roell
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany; NeuroImaging Core Unit Munich, LMU University Hospital, LMU Munich, Munich, Germany
| | - Genc Hasanaj
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany; Evidence-Based Psychiatry and Psychotherapy, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Simiao Xu
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Joanna Moussiopoulou
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Siegfried Priglinger
- Department of Ophthalmology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Christoph Kern
- Department of Ophthalmology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Eva C Schulte
- Institute of Psychiatric Phenomics and Genomics, LMU Munich, Munich, Germany; Institute of Human Genetics, University Hospital, Faculty of Medicine, University of Bonn, Bonn, Germany; Department of Psychiatry and Psychotherapy, University Hospital, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Alkomiet Hasan
- Department of Psychiatry, Psychotherapy, and Psychosomatics, Faculty of Medicine, University of Augsburg, Augsburg, Germany; German Center for Mental Health, partner site Munich-Augsburg, Germany
| | - Oliver Pogarell
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany
| | - Peter Falkai
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany; Max Planck Institute of Psychiatry, Munich, Germany; German Center for Mental Health, partner site Munich-Augsburg, Germany
| | - Andrea Schmitt
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany; Max Planck Institute of Psychiatry, Munich, Germany; German Center for Mental Health, partner site Munich-Augsburg, Germany; Laboratory of Neurosciences (LIM-27), Institute of Psychiatry, University of São Paulo, São Paulo, Brazil
| | - Benedikt Schworm
- Department of Ophthalmology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Elias Wagner
- Evidence-Based Psychiatry and Psychotherapy, Faculty of Medicine, University of Augsburg, Augsburg, Germany; Department of Psychiatry, Psychotherapy, and Psychosomatics, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | - Daniel Keeser
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany; NeuroImaging Core Unit Munich, LMU University Hospital, LMU Munich, Munich, Germany; Munich Center for Neurosciences, LMU Munich, Planegg-Martinsried, Germany
| | - Florian J Raabe
- Department of Psychiatry and Psychotherapy, LMU University Hospital, LMU Munich, Munich, Germany; Max Planck Institute of Psychiatry, Munich, Germany.
| |
Collapse
|
9
|
Skelin L, Racetin A, Kelam N, Ogorevc M, Znaor L, Saraga-Babić M, Filipović N, Katsuyama Y, Pogorelić Z, Vukojević K. Connexin Expression Is Altered in the Eye Development of Yotari Mice: A Preliminary Study. Biomolecules 2024; 14:1174. [PMID: 39334940 PMCID: PMC11430515 DOI: 10.3390/biom14091174] [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: 08/20/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
This study aimed to explore how Dab1 functional silencing influences the expression patterns of different connexins in the developing yotari (yot) mice eyes as potential determinants of retinogenesis. Using immunofluorescence staining, the protein expression of Dab1, Reelin, and connexin 37, 40, 43, and 45 (Cx37, Cx40, Cx43, and Cx45) in the wild-type (wt) and yot eyes at embryonic days 13.5 and 15.5 (E13.5 and E15.5) were analyzed. Different expression patterns of Cx37 were seen between the wt and yot groups. The highest fluorescence intensity of Cx37 was observed in the yot animals at E15.5. Cx40 had higher expression at the E13.5 when differentiation of retinal layers was still beginning, whereas it decreased at the E15.5 when differentiation was at the advanced stage. Higher expression of Cx43 was found in the yot group at both time points. Cx45 was predominantly expressed at E13.5 in both groups. Our results reveal the altered expression of connexins during retinogenesis in yot mice and their potential involvement in retinal pathology, where they might serve as prospective therapeutic targets.
Collapse
Affiliation(s)
- Ljubica Skelin
- Clinical Department of Ophthalmology, University Hospital of Split, 21000 Split, Croatia
| | - Anita Racetin
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Nela Kelam
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Marin Ogorevc
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Ljubo Znaor
- Clinical Department of Ophthalmology, University Hospital of Split, 21000 Split, Croatia
- Department of Ophthalmology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Mirna Saraga-Babić
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Natalija Filipović
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| | - Yu Katsuyama
- Department of Anatomy, Shiga University of Medical Science, Otsu 520-2192, Japan
| | - Zenon Pogorelić
- Department of Pediatric Surgery, Split University Hospital, 21000 Split, Croatia
| | - Katarina Vukojević
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
- Center for Translational Research in Biomedicine, University of Split School of Medicine, Šoltanska 2A, 21000 Split, Croatia
| |
Collapse
|
10
|
Luo Z, Xu Y, Xiong X, Huang S, Alimu S, Cui J, Xu K, Tsui CK, Fan S, Cui K, Yu S, Liang X. Characterization of Vortex Vein Drainage System in Healthy Individuals Imaged by Ultra-Widefield Optical Coherence Tomography Angiography. Transl Vis Sci Technol 2024; 13:19. [PMID: 39292469 PMCID: PMC11412622 DOI: 10.1167/tvst.13.9.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024] Open
Abstract
Purpose The purpose of this study was to investigate the choroidal characteristics of vortex vein (VV) drainage systems in healthy individuals using ultra-widefield optical coherence tomography angiography. Methods The mean choroidal thickness (ChT) and choroidal vascularity index (CVI) of each VV quadrant (24 × 20 mm2 scan mode; superotemporal [ST], superonasal [SN], inferonasal [IN], and inferotemporal [IT] quadrants) were calculated. Furthermore, intervortex venous anastomosis (IVA) was classified into temporal, superior, inferior, and nasal types. Results A total of 207 healthy eyes were analyzed to find that the ST quadrant had the thickest choroidal layer and highest CVI (all P < 0.05). Among the four VV drainage quadrants, the mean ChT and CVI decreased in the sequence of ST, SN, IT, and IN (all P < 0.05). Moreover, men had a higher CVI than women in all VV quadrants (all P < 0.05). IVA was observed in all VV quadrants of 91 eyes (43.96%), and in the macular region of 33 eyes (15.94%). Conclusions The ST drainage system was identified as the preferred VV drainage route in healthy eyes. Among the four VV drainage quadrants, the drainage system adhered to the ST-SN-IT-IN order of descending perfusion. Furthermore, age- and sex-related differences were noted in the choroidal VV drainage systems of healthy eyes. Additionally, almost half of the healthy eyes had IVA in their choroidal vessel networks. Translational Relevance The VV drainage system may be considered a novel imaging biomarker for ocular diseases.
Collapse
Affiliation(s)
- Zhonghua Luo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Xiaomei Xiong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Shengsong Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Subinuer Alimu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Jinli Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Kun Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Ching-Kit Tsui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Shuxin Fan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Kaixuan Cui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Shanshan Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangdong Provincial Clinical Research Center for Ocular Diseases, Guangzhou, China
| |
Collapse
|
11
|
You Y, Fu Y, Li L, Zhang Z, Jia S, Lu S, Ren W, Liu Y, Xu Y, Liu X, Jiang F, Peng G, Sampath Kumar A, Ritchie ME, Liu X, Tian L. Systematic comparison of sequencing-based spatial transcriptomic methods. Nat Methods 2024; 21:1743-1754. [PMID: 38965443 PMCID: PMC11399101 DOI: 10.1038/s41592-024-02325-3] [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: 03/04/2024] [Accepted: 05/29/2024] [Indexed: 07/06/2024]
Abstract
Recent developments of sequencing-based spatial transcriptomics (sST) have catalyzed important advancements by facilitating transcriptome-scale spatial gene expression measurement. Despite this progress, efforts to comprehensively benchmark different platforms are currently lacking. The extant variability across technologies and datasets poses challenges in formulating standardized evaluation metrics. In this study, we established a collection of reference tissues and regions characterized by well-defined histological architectures, and used them to generate data to compare 11 sST methods. We highlighted molecular diffusion as a variable parameter across different methods and tissues, significantly affecting the effective resolutions. Furthermore, we observed that spatial transcriptomic data demonstrate unique attributes beyond merely adding a spatial axis to single-cell data, including an enhanced ability to capture patterned rare cell states along with specific markers, albeit being influenced by multiple factors including sequencing depth and resolution. Our study assists biologists in sST platform selection, and helps foster a consensus on evaluation standards and establish a framework for future benchmarking efforts that can be used as a gold standard for the development and benchmarking of computational tools for spatial transcriptomic analysis.
Collapse
Affiliation(s)
- Yue You
- Guangzhou National Laboratory, Guangzhou, China
| | - Yuting Fu
- School of Life Sciences, Westlake University, Hangzhou, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Westlake Institute for Advanced Study, Hangzhou, China
| | - Lanxiang Li
- Guangzhou National Laboratory, Guangzhou, China
| | | | - Shikai Jia
- School of Life Sciences, Westlake University, Hangzhou, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Westlake Institute for Advanced Study, Hangzhou, China
| | - Shihong Lu
- Guangzhou National Laboratory, Guangzhou, China
| | - Wenle Ren
- Guangzhou National Laboratory, Guangzhou, China
| | - Yifang Liu
- School of Life Sciences, Westlake University, Hangzhou, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Westlake Institute for Advanced Study, Hangzhou, China
| | - Yang Xu
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Xiaojing Liu
- School of Life Sciences, Westlake University, Hangzhou, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Westlake Institute for Advanced Study, Hangzhou, China
| | - Fuqing Jiang
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, University of Chinese Academy of Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Guangdun Peng
- Center for Cell Lineage and Development, CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, University of Chinese Academy of Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| | - Abhishek Sampath Kumar
- Department of Stem Cell and Regenerative Biology, Harvard University. Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Matthew E Ritchie
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Xiaodong Liu
- School of Life Sciences, Westlake University, Hangzhou, China.
- Research Center for Industries of the Future, Westlake University, Hangzhou, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- Westlake Institute for Advanced Study, Hangzhou, China.
| | - Luyi Tian
- Guangzhou National Laboratory, Guangzhou, China.
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
12
|
Bouchikh-El Jarroudi R, Valentín-Bravo FJ, Calatayud-Riera M, Ruiz-Bilbao S. Retinal stroke: A call to action for awareness and treatment of ocular vascular emergencies. ARCHIVOS DE LA SOCIEDAD ESPANOLA DE OFTALMOLOGIA 2024; 99:363-364. [PMID: 38670259 DOI: 10.1016/j.oftale.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 03/17/2024] [Indexed: 04/28/2024]
Affiliation(s)
- R Bouchikh-El Jarroudi
- Servicio de Oftalmología, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain; Unidad de Oftalmología Basada en Evidencias (Oftalmoevidencia), Scientia Clinical and Epidemiological Research Institute, Trujillo, Peru.
| | - F J Valentín-Bravo
- Servicio de Oftalmología, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain; Institut Català de la Retina (ICR), Barcelona, Spain
| | - M Calatayud-Riera
- Servicio de Oftalmología, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - S Ruiz-Bilbao
- Servicio de Oftalmología, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| |
Collapse
|
13
|
Rosmus DD, Koch J, Hausmann A, Chiot A, Arnhold F, Masuda T, Kierdorf K, Hansen SM, Kuhrt H, Fröba J, Wolf J, Boneva S, Gericke M, Ajami B, Prinz M, Lange C, Wieghofer P. Redefining the ontogeny of hyalocytes as yolk sac-derived tissue-resident macrophages of the vitreous body. J Neuroinflammation 2024; 21:168. [PMID: 38961498 PMCID: PMC11223341 DOI: 10.1186/s12974-024-03110-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: 01/26/2024] [Accepted: 04/22/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND The eye is a highly specialized sensory organ which encompasses the retina as a part of the central nervous system, but also non-neural compartments such as the transparent vitreous body ensuring stability of the eye globe and a clear optical axis. Hyalocytes are the tissue-resident macrophages of the vitreous body and are considered to play pivotal roles in health and diseases of the vitreoretinal interface, such as proliferative vitreoretinopathy or diabetic retinopathy. However, in contrast to other ocular macrophages, their embryonic origin as well as the extent to which these myeloid cells might be replenished by circulating monocytes remains elusive. RESULTS In this study, we combine transgenic reporter mice, embryonic and adult fate mapping approaches as well as parabiosis experiments with multicolor immunofluorescence labeling and confocal laser-scanning microscopy to comprehensively characterize the murine hyalocyte population throughout development and in adulthood. We found that murine hyalocytes express numerous well-known myeloid cell markers, but concomitantly display a distinct immunophenotype that sets them apart from retinal microglia. Embryonic pulse labeling revealed a yolk sac-derived origin of murine hyalocytes, whose precursors seed the developing eye prenatally. Finally, postnatal labeling and parabiosis established the longevity of hyalocytes which rely on Colony Stimulating Factor 1 Receptor (CSF1R) signaling for their maintenance, independent of blood-derived monocytes. CONCLUSION Our study identifies hyalocytes as long-living progeny of the yolk sac hematopoiesis and highlights their role as integral members of the innate immune system of the eye. As a consequence of their longevity, immunosenescence processes may culminate in hyalocyte dysfunction, thereby contributing to the development of vitreoretinal diseases. Therefore, myeloid cell-targeted therapies that convey their effects through the modification of hyalocyte properties may represent an interesting approach to alleviate the burden imposed by diseases of the vitreoretinal interface.
Collapse
Affiliation(s)
- Dennis-Dominik Rosmus
- Institute of Anatomy, Leipzig University, 04103, Leipzig, Germany
- Cellular Neuroanatomy, Institute of Theoretical Medicine, Augsburg University, Universitätsstrasse 2, 86159, Augsburg, Germany
| | - Jana Koch
- Cellular Neuroanatomy, Institute of Theoretical Medicine, Augsburg University, Universitätsstrasse 2, 86159, Augsburg, Germany
- Institute of Neuropathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Annika Hausmann
- Institute of Neuropathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Aude Chiot
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, 97239, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Franz Arnhold
- Institute of Anatomy, Leipzig University, 04103, Leipzig, Germany
| | - Takahiro Masuda
- Institute of Neuropathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Division of Molecular Neuroimmunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582, Japan
| | - Katrin Kierdorf
- Institute of Neuropathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Centre for Integrative Biological Signalling Studies, University of Freiburg, 79106, Freiburg, Germany
- Centre for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Stefanie Marie Hansen
- Institute of Neuropathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Heidrun Kuhrt
- Institute of Anatomy, Leipzig University, 04103, Leipzig, Germany
| | - Janine Fröba
- Institute of Anatomy, Leipzig University, 04103, Leipzig, Germany
| | - Julian Wolf
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Molecular Surgery Laboratory, Stanford University, Palo Alto, CA, 94304, USA
- Department of Ophthalmology, Byers Eye Institute, Stanford University, Palo Alto, CA, 94304, USA
| | - Stefaniya Boneva
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
| | - Martin Gericke
- Institute of Anatomy, Leipzig University, 04103, Leipzig, Germany
| | - Bahareh Ajami
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, OR, 97239, USA
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Marco Prinz
- Institute of Neuropathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79106, Freiburg, Germany
| | - Clemens Lange
- Eye Center, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany
- Ophtha Lab, Department of Ophthalmology, St. Franziskus Hospital, 48145, Münster, Germany
| | - Peter Wieghofer
- Institute of Anatomy, Leipzig University, 04103, Leipzig, Germany.
- Cellular Neuroanatomy, Institute of Theoretical Medicine, Augsburg University, Universitätsstrasse 2, 86159, Augsburg, Germany.
- Institute of Neuropathology, Medical Center, Faculty of Medicine, University of Freiburg, 79106, Freiburg, Germany.
| |
Collapse
|
14
|
De S, Gangwar A. Unveiling the Impact of Maternal Hyperthermia in the Late First Trimester: A Case Report of Anterior Esthetic Rehabilitation Utilizing Heterodontic Biologic Posts. Cureus 2024; 16:e64922. [PMID: 39156431 PMCID: PMC11330640 DOI: 10.7759/cureus.64922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 07/18/2024] [Indexed: 08/20/2024] Open
Abstract
The perinatal maternal environment is important for the normal development of the fetus. Epigenetic modifications that influence developmental control genes and signalling pathways for proper fetal development have been associated with maternal illnesses brought on by viruses, bacteria, or even parasitic protozoa. It is crucial to provide details on the onset, length, and timing of the mother's fever because these factors may influence the kind of certain abnormalities. Although fever is a primarily benign disease, it has been linked to negative health outcomes in children and has occasionally resulted in a substantial referral to critical care. This case report presents a 15-year-old female patient with repaired cleft palate and tetralogy of Fallot (TOF) who approached for esthetic rehabilitation of lower anterior teeth. The teeth (31, 32, 43) were tender on percussion. Radiographic evaluation showed the presence of periapical radiolucency. The root canal procedure was performed under local anaesthesia, and the supernumerary maxillary teeth were extracted. After cleaning and disinfecting, these teeth were used as biologic posts with respect to 32 and 33. A follow-up examination was performed after 12 months. The results of this case indicate that using autologous heterodontic biologic posts can lead to a favourable outcome.
Collapse
Affiliation(s)
- Sriparna De
- Pediatric Dentistry, Institute of Dental Sciences, Bareilly, Bareilly, IND
| | - Anshul Gangwar
- Pediatric Dentistry, Institute of Dental Sciences, Bareilly, Bareilly, IND
| |
Collapse
|
15
|
Mukhamadiarov RI, Ciarchi M, Olmeda F, Rulands S. Clonal dynamics of surface-driven growing tissues. Phys Rev E 2024; 109:064407. [PMID: 39021023 DOI: 10.1103/physreve.109.064407] [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: 12/15/2023] [Accepted: 05/17/2024] [Indexed: 07/20/2024]
Abstract
The self-organization of cells into complex tissues relies on a tight coordination of cell behavior. Identifying the cellular processes driving tissue growth is key to understanding the emergence of tissue forms and devising targeted therapies for aberrant growth, such as in cancer. Inferring the mode of tissue growth, whether it is driven by cells on the surface or by cells in the bulk, is possible in cell culture experiments but difficult in most tissues in living organisms (in vivo). Genetic tracing experiments, where a subset of cells is labeled with inheritable markers, have become important experimental tools to study cell fate in vivo. Here we show that the mode of tissue growth is reflected in the size distribution of the progeny of marked cells. To this end, we derive the clone size distributions using analytical calculations in the limit of negligible cell migration and cell death, and we test our predictions with an agent-based stochastic sampling technique. We show that for surface-driven growth the clone size distribution takes a characteristic power-law form with an exponent determined by fluctuations of the tissue surface. Our results propose a possible way of determining the mode of tissue growth from genetic tracing experiments.
Collapse
|
16
|
Eker S, Karaküçük Y, Gumus H. Assessment of macular microcirculation in patients with multiple sclerosis by swept-source optical coherence tomography angiography. MARMARA MEDICAL JOURNAL 2024; 37:192-197. [DOI: 10.5472/marumj.1487767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Objective: To investigate the changes in the retinal microcirculation in multiple sclerosis (MS) patients by swept-source optical
coherence tomography angiography (SS-OCTA).
Patients and Methods: Thirty-seven patients with relapsing-remitting MS and 40 healthy volunteers were included into this crosssectional
study. Clinical history, Expanded Disability Status Scale and duration of MS were collected. SS-OCTA by deep range imaging
(DRI) OCT measurements were performed on all subjects. Macular perfusion parameters including superficial and deep foveal
avascular zones (FAZs, FAZd, respectively) (%), vascular densities of superficial capillary plexus (SCP) (%), deep capillary plexus
(DCP) (%) and choriocapillaris (CC) (%) were compared with healthy subjects.
Results: Vascular densities of SCP, DCP and CC were found to be statistically lower in the study group compared to the control group
(p = 0.02, p = 0.03, p = 0.03, respectively). FAZs and FAZd, areas were significantly higher in the study group (p = 0.02, p = 0.02,
respectively). Central macular thickness and subfoveal choroidal thickness were significantly lower than in the control group (p =
0.015, p = 0.047, respectively).
Conclusion: Evaluation of retinal blood flow in patients with MS is useful both for understanding the physiopathology of the disease
and in the clinical follow-up.
Collapse
|
17
|
Huniadi M, Nosálová N, Almášiová V, Horňáková Ľ, Valenčáková A, Hudáková N, Cizkova D. Three-Dimensional Cultivation a Valuable Tool for Modelling Canine Mammary Gland Tumour Behaviour In Vitro. Cells 2024; 13:695. [PMID: 38667310 PMCID: PMC11049302 DOI: 10.3390/cells13080695] [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: 02/29/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Cell cultivation has been one of the most popular methods in research for decades. Currently, scientists routinely use two-dimensional (2D) and three-dimensional (3D) cell cultures of commercially available cell lines and primary cultures to study cellular behaviour, responses to stimuli, and interactions with their environment in a controlled laboratory setting. In recent years, 3D cultivation has gained more attention in modern biomedical research, mainly due to its numerous advantages compared to 2D cultures. One of the main goals where 3D culture models are used is the investigation of tumour diseases, in both animals and humans. The ability to simulate the tumour microenvironment and design 3D masses allows us to monitor all the processes that take place in tumour tissue created not only from cell lines but directly from the patient's tumour cells. One of the tumour types for which 3D culture methods are often used in research is the canine mammary gland tumour (CMT). The clinically similar profile of the CMT and breast tumours in humans makes the CMT a suitable model for studying the issue not only in animals but also in women.
Collapse
Affiliation(s)
- Mykhailo Huniadi
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovakia; (M.H.); (N.N.); (Ľ.H.); (A.V.); (N.H.)
| | - Natália Nosálová
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovakia; (M.H.); (N.N.); (Ľ.H.); (A.V.); (N.H.)
| | - Viera Almášiová
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovakia;
| | - Ľubica Horňáková
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovakia; (M.H.); (N.N.); (Ľ.H.); (A.V.); (N.H.)
| | - Alexandra Valenčáková
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovakia; (M.H.); (N.N.); (Ľ.H.); (A.V.); (N.H.)
| | - Nikola Hudáková
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovakia; (M.H.); (N.N.); (Ľ.H.); (A.V.); (N.H.)
| | - Dasa Cizkova
- Small Animal Clinic, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovakia; (M.H.); (N.N.); (Ľ.H.); (A.V.); (N.H.)
| |
Collapse
|
18
|
Piñon-Teal WL, Ogilvie JM. G protein-coupled estrogen receptor expression in postnatal developing mouse retina. FRONTIERS IN OPHTHALMOLOGY 2024; 4:1331298. [PMID: 38984123 PMCID: PMC11182193 DOI: 10.3389/fopht.2024.1331298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/13/2024] [Indexed: 07/11/2024]
Abstract
Introduction Estrogen has emerged as a multifaceted signaling molecule in the retina, playing an important role in neural development and providing neuroprotection in adults. It interacts with two receptor types: classical estrogen receptors (ERs) alpha and beta, and G protein-coupled estrogen receptor (Gper). Gper differs from classical ERs in structure, localization, and signaling. Here we provide the first report of the temporal and spatial properties of Gper transcript and protein expression in the developing and mature mouse retina. Methods We applied qRT-PCR to determine Gper transcript expression in wild type mouse retina from P0-P21. Immunohistochemistry and Western blot were used to determine Gper protein expression and localization at the same time points. Results Gper expression showed a 6-fold increase during postnatal development, peaking at P14. Relative total Gper expression exhibited a significant decrease during retinal development, although variations emerged in the timing of changes among different forms of the protein. Gper immunoreactivity was seen in retinal ganglion cells (RGCs) throughout development and also in somas in the position of horizontal cells at early time points. Immunoreactivity was observed in the cytoplasm and Golgi at all time points, in the nucleus at early time points, and in RGC axons as the retina matured. Discussion In conclusion, our study illuminates the spatial and temporal expression patterns of Gper in the developing mouse retina and provides a vital foundation for further investigations into the role of Gper in retinal development and degeneration.
Collapse
Affiliation(s)
| | - Judith Mosinger Ogilvie
- Department of Biology, Saint Louis University, St. Louis, MO, United States
- Institute for Translational Neuroscience, Saint Louis University, St. Louis, MO, United States
| |
Collapse
|
19
|
Zheng Y, Stormo GD, Chen S. Aberrant homeodomain-DNA cooperative dimerization underlies distinct developmental defects in two dominant CRX retinopathy models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.12.584677. [PMID: 38559186 PMCID: PMC10979960 DOI: 10.1101/2024.03.12.584677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Paired-class homeodomain transcription factors (HD TFs) play essential roles in vertebrate development, and their mutations are linked to human diseases. One unique feature of paired-class HD is cooperative dimerization on specific palindrome DNA sequences. Yet, the functional significance of HD cooperative dimerization in animal development and its dysregulation in diseases remain elusive. Using the retinal TF Cone-rod Homeobox (CRX) as a model, we have studied how blindness-causing mutations in the paired HD, p.E80A and p.K88N, alter CRX's cooperative dimerization, lead to gene misexpression and photoreceptor developmental deficits in dominant manners. CRXE80A maintains binding at monomeric WT CRX motifs but is deficient in cooperative binding at dimeric motifs. CRXE80A's cooperativity defect impacts the exponential increase of photoreceptor gene expression in terminal differentiation and produces immature, non-functional photoreceptors in the CrxE80A retinas. CRXK88N is highly cooperative and localizes to ectopic genomic sites with strong enrichment of dimeric HD motifs. CRXK88N's altered biochemical properties disrupt CRX's ability to direct dynamic chromatin remodeling during development to activate photoreceptor differentiation programs and silence progenitor programs. Our study here provides in vitro and in vivo molecular evidence that paired-class HD cooperative dimerization regulates neuronal development and dysregulation of cooperative binding contributes to severe dominant blinding retinopathies.
Collapse
Affiliation(s)
- Yiqiao Zheng
- Molecular Genetics and Genomics Graduate Program, Division of Biological and Biomedical Sciences, Washington University in St Louis, Saint Louis, Missouri, 63110, USA
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis, Saint Louis, Missouri, 63110, USA
| | - Gary D. Stormo
- Department of Genetics, Washington University in St Louis, Saint Louis, Missouri, 63110, USA
| | - Shiming Chen
- Department of Ophthalmology and Visual Sciences, Washington University in St Louis, Saint Louis, Missouri, 63110, USA
- Department of Developmental Biology, Washington University in St Louis, Saint Louis, Missouri, 63110, USA
| |
Collapse
|
20
|
Xiong S, He J, Qiu H, van Gestel CAM, He E, Qiao Z, Cao L, Li J, Chen G. Maternal exposure to polystyrene nanoplastics causes defective retinal development and function in progeny mice by disturbing metabolic profiles. CHEMOSPHERE 2024; 352:141513. [PMID: 38387657 DOI: 10.1016/j.chemosphere.2024.141513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/23/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Microplastics (MPs) and nanoplastics (NPs) are widely spreading in our living environment, accumulating in the human body and potentially threating human health. The retina, which is a terminally differentiated extension of the central nervous system, is essential for the visual system. However, the effects and molecular mechanisms of MPs/NPs on retina development and function are still unclear. Here, we investigated the effects and modes of action of polystyrene NPs (PS-NPs) on the retina using mice as a mammalian model species. Maternal PS-NP exposure (100 nm) at an environmentally realistic concentration of 10 mg L-1 (or 2.07 *1010 particles mL-1) via drinking water from the first day of pregnancy till the end of lactation (21 days after birth) caused defective neural retinal development in the neonatal mice, by depositing in the retinal tissue and reducing the number of retinal ganglion cells and bipolar cells. Exposure to PS-NPs retarded retinal vascular development, while abnormal electroretinogram (ERG) responses and an increased level of oxidative stress were also observed in the retina of the progeny mice after maternal PS-NP exposure. Metabolomics showed significant dysregulation of amino acids that are pivotal to neuron retinal function, such as glutamate, aspartate, alanine, glycine, serine, threonine, taurine, and serotonin. Transcriptomics identified significantly dysregulated genes, which were enriched in processes of angiogenesis, visual system development and lens development. Regulatory analysis showed that Fos gene mediated pathways could be a potential key target for PS-NP exposure in retinal development and function. Our study revealed that maternal exposure to PS-NPs generated detrimental effects on retinal development and function in progeny mice, offering new insights into the visual toxicity of PS-NPs.
Collapse
Affiliation(s)
- Shiyi Xiong
- Shanghai Key Laboratory of Maternal Fetal Medicine, Department of Fetal Medicine and Prenatal Diagnosis Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China
| | - Jincan He
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hao Qiu
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Cornelis A M van Gestel
- Amsterdam Institute for Life and Environment (A-LIFE), Faculty of Science, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, the Netherlands
| | - ErKai He
- School of Geographic Sciences, East China Normal University, Shanghai, 200241, China
| | - Zhengdong Qiao
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Liang Cao
- Department of Ophthalmology, Shanghai International Medical Center, Shanghai, China
| | - Jing Li
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Guangquan Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Department of Fetal Medicine and Prenatal Diagnosis Center, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 201204, China.
| |
Collapse
|
21
|
Replogle MR, Thompson S, Reis LM, Semina EV. A De Novo Noncoding RARB Variant Associated with Complex Microphthalmia Alters a Putative Regulatory Element. Hum Mutat 2024; 2024:6619280. [PMID: 39450403 PMCID: PMC11501074 DOI: 10.1155/2024/6619280] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Retinoic acid receptor beta (RARB) is a transcriptional regulator crucial for coordinating retinoic acid- (RA-) mediated morphogenic movements, cell growth, and differentiation during eye development. Loss- or gain-of-function RARB coding variants have been associated with microphthalmia, coloboma, and anterior segment defects. We identified a de novo variant c.157+1895G>A located within a conserved region (CR1) in the first intron of RARB in an individual with complex microphthalmia and significant global developmental delay. Based on the phenotypic overlap, we further investigated the possible effects of the variant on mRNA splicing and/or transcriptional regulation through in silico and functional studies. In silico analysis identified the possibility of alternative splicing, suggested by one out of three (HSF, SpliceAI, and MaxEntScan) splicing prediction programs, and a strong indication of regulatory function based on publicly available DNase hypersensitivity, histone modification, chromatin folding, and ChIP-seq data sets. Consistent with the predictions of SpliceAI and MaxEntScan, in vitro minigene assays showed no effect on RARB mRNA splicing. Evaluation of CR1 for a regulatory role using luciferase reporter assays in human lens epithelial cells demonstrated a significant increase in the activity of the RARB promoter in the presence of wild-type CR1. This activity was further significantly increased in the presence of CR1 carrying the c.157+1895G>A variant, suggesting that the variant may promote RARB overexpression in human cells. Induction of RARB overexpression in human lens epithelial cells resulted in increased cell proliferation and elevated expression of FOXC1, a known downstream target of RA signaling and a transcription factor whose down- and upregulation is associated with ocular phenotypes overlapping the RARB spectrum. These results support a regulatory role for the CR1 element and suggest that the de novo c.157+1895G>A variant affecting this region may alter the proper regulation of RARB and, as a result, its downstream genes, possibly leading to abnormal development.
Collapse
Affiliation(s)
- Maria R. Replogle
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Samuel Thompson
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Linda M. Reis
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Elena V. Semina
- Department of Ophthalmology and Visual Sciences, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Pediatrics and Children’s Research Institute, Medical College of Wisconsin and Children’s Hospital of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
22
|
Wu D, Khan FA, Zhang K, Pandupuspitasari NS, Negara W, Guan K, Sun F, Huang C. Retinoic acid signaling in development and differentiation commitment and its regulatory topology. Chem Biol Interact 2024; 387:110773. [PMID: 37977248 DOI: 10.1016/j.cbi.2023.110773] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/11/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023]
Abstract
Retinoic acid (RA), the derivative of vitamin A/retinol, is a signaling molecule with important implications in health and disease. It is a well-known developmental morphogen that functions mainly through the transcriptional activity of nuclear RA receptors (RARs) and, uncommonly, through other nuclear receptors, including peroxisome proliferator-activated receptors. Intracellular RA is under spatiotemporally fine-tuned regulation by synthesis and degradation processes catalyzed by retinaldehyde dehydrogenases and P450 family enzymes, respectively. In addition to dictating the transcription architecture, RA also impinges on cell functioning through non-genomic mechanisms independent of RAR transcriptional activity. Although RA-based differentiation therapy has achieved impressive success in the treatment of hematologic malignancies, RA also has pro-tumor activity. Here, we highlight the relevance of RA signaling in cell-fate determination, neurogenesis, visual function, inflammatory responses and gametogenesis commitment. Genetic and post-translational modifications of RAR are also discussed. A better understanding of RA signaling will foster the development of precision medicine to improve the defects caused by deregulated RA signaling.
Collapse
Affiliation(s)
- Di Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | - Faheem Ahmed Khan
- Research Center for Animal Husbandry, National Research and Innovation Agency, Jakarta Pusat, 10340, Indonesia
| | - Kejia Zhang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China
| | | | - Windu Negara
- Research Center for Animal Husbandry, National Research and Innovation Agency, Jakarta Pusat, 10340, Indonesia
| | - Kaifeng Guan
- School of Advanced Agricultural Sciences, Peking University, Beijing, 100871, China.
| | - Fei Sun
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| | - Chunjie Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong, 226001, China.
| |
Collapse
|
23
|
Zhang K, Cai W, Hu L, Chen S. Generating Retinas through Guided Pluripotent Stem Cell Differentiation and Direct Somatic Cell Reprogramming. Curr Stem Cell Res Ther 2024; 19:1251-1262. [PMID: 37807418 DOI: 10.2174/011574888x255496230923164547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/25/2023] [Accepted: 08/28/2023] [Indexed: 10/10/2023]
Abstract
Retinal degeneration diseases affect millions of people worldwide but are among the most difficult eye diseases to cure. Studying the mechanisms and developing new therapies for these blinding diseases requires researchers to have access to many retinal cells. In recent years there has been substantial advances in the field of biotechnology in generating retinal cells and even tissues in vitro, either through programmed sequential stem cell differentiation or direct somatic cell lineage reprogramming. The resemblance of these in vitro-generated retinal cells to native cells has been increasingly utilized by researchers. With the help of these in vitro retinal models, we now have a better understanding of human retinas and retinal diseases. Furthermore, these in vitro-generated retinal cells can be used as donor cells which solves a major hurdle in the development of cell replacement therapy for retinal degeneration diseases, while providing a promising option for patients suffering from these diseases. In this review, we summarize the development of pluripotent stem cell-to-retinal cell differentiation methods, the recent advances in generating retinal cells through direct somatic cell reprogramming, and the translational applications of retinal cells generated in vitro. Finally, we discuss the limitations of the current protocols and possible future directions for improvement.
Collapse
Affiliation(s)
- Ke Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510623, China
| | - Wenwen Cai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510623, China
| | - Leyi Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510623, 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, 510623, China
| |
Collapse
|
24
|
Dubaic M, Peskova L, Hampl M, Weissova K, Celiker C, Shylo NA, Hruba E, Kavkova M, Zikmund T, Weatherbee SD, Kaiser J, Barta T, Buchtova M. Role of ciliopathy protein TMEM107 in eye development: insights from a mouse model and retinal organoid. Life Sci Alliance 2023; 6:e202302073. [PMID: 37863656 PMCID: PMC10589122 DOI: 10.26508/lsa.202302073] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 10/22/2023] Open
Abstract
Primary cilia are cellular surface projections enriched in receptors and signaling molecules, acting as signaling hubs that respond to stimuli. Malfunctions in primary cilia have been linked to human diseases, including retinopathies and ocular defects. Here, we focus on TMEM107, a protein localized to the transition zone of primary cilia. TMEM107 mutations were found in patients with Joubert and Meckel-Gruber syndromes. A mouse model lacking Tmem107 exhibited eye defects such as anophthalmia and microphthalmia, affecting retina differentiation. Tmem107 expression during prenatal mouse development correlated with phenotype occurrence, with enhanced expression in differentiating retina and optic stalk. TMEM107 deficiency in retinal organoids resulted in the loss of primary cilia, down-regulation of retina-specific genes, and cyst formation. Knocking out TMEM107 in human ARPE-19 cells prevented primary cilia formation and impaired response to Smoothened agonist treatment because of ectopic activation of the SHH pathway. Our data suggest TMEM107 plays a crucial role in early vertebrate eye development and ciliogenesis in the differentiating retina.
Collapse
Affiliation(s)
- Marija Dubaic
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Lucie Peskova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marek Hampl
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Kamila Weissova
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czech Republic
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Canan Celiker
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Natalia A Shylo
- Department of Genetics, Yale University, School of Medicine, New Haven, CT, USA
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Eva Hruba
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czech Republic
| | - Michaela Kavkova
- CEITEC - Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Tomas Zikmund
- CEITEC - Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Scott D Weatherbee
- Department of Genetics, Yale University, School of Medicine, New Haven, CT, USA
- Biology Department, Fairfield University, Fairfield, CT, USA
| | - Jozef Kaiser
- CEITEC - Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Tomas Barta
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czech Republic
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marcela Buchtova
- Laboratory of Molecular Morphogenesis, Institute of Animal Physiology and Genetics, Czech Academy of Sciences, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| |
Collapse
|
25
|
Abdalkader RK, Fujita T. Corneal epithelium models for safety assessment in drug development: Present and future directions. Exp Eye Res 2023; 237:109697. [PMID: 37890755 DOI: 10.1016/j.exer.2023.109697] [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: 06/30/2022] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 10/29/2023]
Abstract
The human corneal epithelial barrier plays a crucial role in drug testing studies, including drug absorption, distribution, metabolism, and excretion (ADME), as well as toxicity testing during the preclinical stages of drug development. However, despite the valuable insights gained from animal and current in vitro models, there remains a significant discrepancy between preclinical drug predictions and actual clinical outcomes. Additionally, there is a growing emphasis on adhering to the 3R principles (refine, reduce, replace) to minimize the use of animals in testing. To tackle these challenges, there is a rising demand for alternative in vitro models that closely mimic the human corneal epithelium. Recently, remarkable advancements have been made in two key areas: microphysiological systems (MPS) or organs-on-chips (OoCs), and stem cell-derived organoids. These cutting-edge platforms integrate four major disciplines: stem cells, microfluidics, bioprinting, and biosensing technologies. This integration holds great promise in developing powerful and biomimetic models of the human cornea.
Collapse
Affiliation(s)
- Rodi Kado Abdalkader
- Ritsumeikan Global Innovation Research Organization (R-GIRO), Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan.
| | - Takuya Fujita
- Ritsumeikan Global Innovation Research Organization (R-GIRO), Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan; Department of Pharmaceutical Sciences, Ritsumeikan University, 1-1-1 Noji-Higashi, Kusatsu, Shiga, 525-8577, Japan
| |
Collapse
|
26
|
Springer MS, Emerling CA, Gatesy J. Three Blind Moles: Molecular Evolutionary Insights on the Tempo and Mode of Convergent Eye Degeneration in Notoryctes typhlops (Southern Marsupial Mole) and Two Chrysochlorids (Golden Moles). Genes (Basel) 2023; 14:2018. [PMID: 38002961 PMCID: PMC10671557 DOI: 10.3390/genes14112018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/20/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Golden moles (Chrysochloridae) and marsupial moles (Notoryctidae) are textbook examples of convergent evolution. Both taxa are highly adapted to subterranean lifestyles and have powerful limbs for digging through the soil/sand, ears that are adapted for low-frequency hearing, vestigial eyes that are covered by skin and fur, and the absence of optic nerve connections between the eyes and the brain. The eyes of marsupial moles also lack a lens as well as retinal rods and cones. Two hypotheses have been proposed to account for the greater degeneracy of the eyes of marsupial moles than golden moles. First, marsupial moles may have had more time to adapt to their underground habitat than other moles. Second, the eyes of marsupial moles may have been rapidly and recently vestigialized to (1) reduce the injurious effects of sand getting into the eyes and (2) accommodate the enlargement of lacrimal glands that keep the nasal cavity moist and prevent the entry of sand into the nasal passages during burrowing. Here, we employ molecular evolutionary methods on DNA sequences for 38 eye genes, most of which are eye-specific, to investigate the timing of relaxed selection (=neutral evolution) for different groups of eye-specific genes that serve as proxies for distinct functional components of the eye (rod phototransduction, cone phototransduction, lens/cornea). Our taxon sampling included 12 afrothere species, of which two are golden moles (Amblysomus hottentotus, Chrysochloris asiatica), and 28 marsupial species including two individuals of the southern marsupial mole (Notoryctes typhlops). Most of the sequences were mined from databases, but we also provide new genome data for A. hottentotus and one of the two N. typhlops individuals. Even though the eyes of golden moles are less degenerate than the eyes of marsupial moles, there are more inactivating mutations (e.g., frameshift indels, premature stop codons) in their cone phototransduction and lens/cornea genes than in orthologous genes of the marsupial mole. We estimate that cone phototransduction recovery genes were inactivated first in each group, followed by lens/cornea genes and then cone phototransduction activation genes. All three groups of genes were inactivated earlier in golden moles than in marsupial moles. For the latter, we estimate that lens/cornea genes were inactivated ~17.8 million years ago (MYA) when stem notoryctids were burrowing in the soft soils of Australian rainforests. Selection on phototransduction activation genes was relaxed much later (5.38 MYA), during the early stages of Australia's aridification that produced coastal sand plains and eventually sand dunes. Unlike cone phototransduction activation genes, rod phototransduction activation genes are intact in both golden moles and one of the two individuals of N. typhlops. A second marsupial mole individual has just a single inactivating mutation in one of the rod phototransduction activation genes (PDE6B). One explanation for this result is that some rod phototransduction activation genes are pleiotropic and are expressed in extraocular tissues, possibly in conjunction with sperm thermotaxis.
Collapse
Affiliation(s)
- Mark S. Springer
- Department of Evolution, Ecology, and Organismal Biology, University of California, Riverside, CA 92521, USA
| | | | - John Gatesy
- Division of Vertebrate Zoology, American Museum of Natural History, New York, NY 10024, USA;
| |
Collapse
|
27
|
Agarwal D, Dash N, Mazo KW, Chopra M, Avila MP, Patel A, Wong RM, Jia C, Do H, Cheng J, Chiang C, Jurlina SL, Roshan M, Perry MW, Rho JM, Broyer R, Lee CD, Weinreb RN, Gavrilovici C, Oesch NW, Welsbie DS, Wahlin KJ. Human retinal ganglion cell neurons generated by synchronous BMP inhibition and transcription factor mediated reprogramming. NPJ Regen Med 2023; 8:55. [PMID: 37773257 PMCID: PMC10541876 DOI: 10.1038/s41536-023-00327-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 08/31/2023] [Indexed: 10/01/2023] Open
Abstract
In optic neuropathies, including glaucoma, retinal ganglion cells (RGCs) die. Cell transplantation and endogenous regeneration offer strategies for retinal repair, however, developmental programs required for this to succeed are incompletely understood. To address this, we explored cellular reprogramming with transcription factor (TF) regulators of RGC development which were integrated into human pluripotent stem cells (PSCs) as inducible gene cassettes. When the pioneer factor NEUROG2 was combined with RGC-expressed TFs (ATOH7, ISL1, and POU4F2) some conversion was observed and when pre-patterned by BMP inhibition, RGC-like induced neurons (RGC-iNs) were generated with high efficiency in just under a week. These exhibited transcriptional profiles that were reminiscent of RGCs and exhibited electrophysiological properties, including AMPA-mediated synaptic transmission. Additionally, we demonstrated that small molecule inhibitors of DLK/LZK and GCK-IV can block neuronal death in two pharmacological axon injury models. Combining developmental patterning with RGC-specific TFs thus provided valuable insight into strategies for cell replacement and neuroprotection.
Collapse
Affiliation(s)
- Devansh Agarwal
- Shu Chien-Gene Lay Department of Bioengineering, UC San Diego, La Jolla, CA, USA
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Nicholas Dash
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Kevin W Mazo
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Manan Chopra
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Maria P Avila
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Amit Patel
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Ryan M Wong
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Cairang Jia
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Hope Do
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Jie Cheng
- Department of Ophthalmology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Colette Chiang
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Shawna L Jurlina
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Mona Roshan
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Michael W Perry
- Department of Biological Sciences, UC San Diego, La Jolla, CA, USA
| | - Jong M Rho
- Department of Neurosciences, UC San Diego, La Jolla, CA, USA
| | - Risa Broyer
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Cassidy D Lee
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Robert N Weinreb
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | | | - Nicholas W Oesch
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
- Department of Psychology, UC San Diego, La Jolla, CA, USA
| | - Derek S Welsbie
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA
| | - Karl J Wahlin
- Viterbi Family Department of Ophthalmology & the Shiley Eye Institute, UC San Diego, La Jolla, CA, USA.
| |
Collapse
|
28
|
Beaver D, Limnios IJ. A treatment within sight: challenges in the development of stem cell-derived photoreceptor therapies for retinal degenerative diseases. FRONTIERS IN TRANSPLANTATION 2023; 2:1130086. [PMID: 38993872 PMCID: PMC11235385 DOI: 10.3389/frtra.2023.1130086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 09/07/2023] [Indexed: 07/13/2024]
Abstract
Stem cell therapies can potentially treat various retinal degenerative diseases, including age-related macular degeneration (AMD) and inherited retinal diseases like retinitis pigmentosa. For these diseases, transplanted cells may include stem cell-derived retinal pigmented epithelial (RPE) cells, photoreceptors, or a combination of both. Although stem cell-derived RPE cells have progressed to human clinical trials, therapies using photoreceptors and other retinal cell types are lagging. In this review, we discuss the potential use of human pluripotent stem cell (hPSC)-derived photoreceptors for the treatment of retinal degeneration and highlight the progress and challenges for their efficient production and clinical application in regenerative medicine.
Collapse
Affiliation(s)
- Davinia Beaver
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QL, Australia
| | - Ioannis Jason Limnios
- Clem Jones Centre for Regenerative Medicine, Bond University, Gold Coast, QL, Australia
| |
Collapse
|
29
|
Shi X, Xue Z, Ye K, Yuan J, Zhang Y, Qu J, Su J. Roles of non-coding RNAs in eye development and diseases. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 14:e1785. [PMID: 36849659 DOI: 10.1002/wrna.1785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/17/2022] [Accepted: 02/06/2023] [Indexed: 03/01/2023]
Abstract
The prevalence of ocular disorders is dramatically increasing worldwide, especially those that cause visual impairment and permanent loss of vision, including cataract, glaucoma, age-related macular degeneration, and diabetic retinopathy. Extensive evidence has shown that ncRNAs are key regulators in various biogenesis and biological functions, controlling gene expression related to histogenesis and cell differentiation in ocular tissues. Aberrant expression and function of ncRNA can lead to dysfunction of visual system and mediate progression of eye disorders. Here, we mainly offer an overview of the role of precise modulation of ncRNAs in eye development and function in patients with eye diseases. We also highlight the challenges and future perspectives in conducting ncRNA studies, focusing specifically on the role of ncRNAs that may hold expanded promise for their diagnostic and therapeutic applications in various eye diseases. This article is categorized under: Regulatory RNAs/RNAi/Riboswitches > Regulatory RNAs RNA in Disease and Development > RNA in Disease RNA in Disease and Development > RNA in Development.
Collapse
Affiliation(s)
- Xinrui Shi
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhengbo Xue
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kaicheng Ye
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Yuan
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Zhejiang, China
| | - Yan Zhang
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jia Qu
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Zhejiang, China
| | - Jianzhong Su
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Zhejiang, China
- Institute of PSI Genomics, Zhejiang, China
| |
Collapse
|
30
|
Liu L, Oura S, Markham Z, Hamilton JN, Skory RM, Li L, Sakurai M, Wang L, Pinzon-Arteaga CA, Plachta N, Hon GC, Wu J. Modeling post-implantation stages of human development into early organogenesis with stem-cell-derived peri-gastruloids. Cell 2023; 186:3776-3792.e16. [PMID: 37478861 DOI: 10.1016/j.cell.2023.07.018] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/23/2023]
Abstract
In vitro stem cell models that replicate human gastrulation have been generated, but they lack the essential extraembryonic cells needed for embryonic development, morphogenesis, and patterning. Here, we describe a robust and efficient method that prompts human extended pluripotent stem cells to self-organize into embryo-like structures, termed peri-gastruloids, which encompass both embryonic (epiblast) and extraembryonic (hypoblast) tissues. Although peri-gastruloids are not viable due to the exclusion of trophoblasts, they recapitulate critical stages of human peri-gastrulation development, such as forming amniotic and yolk sac cavities, developing bilaminar and trilaminar embryonic discs, specifying primordial germ cells, initiating gastrulation, and undergoing early neurulation and organogenesis. Single-cell RNA-sequencing unveiled transcriptomic similarities between advanced human peri-gastruloids and primary peri-gastrulation cell types found in humans and non-human primates. This peri-gastruloid platform allows for further exploration beyond gastrulation and may potentially aid in the development of human fetal tissues for use in regenerative medicine.
Collapse
Affiliation(s)
- Lizhong Liu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Seiya Oura
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zachary Markham
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - James N Hamilton
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Robin M Skory
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Leijie Li
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Masahiro Sakurai
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lei Wang
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Carlos A Pinzon-Arteaga
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nicolas Plachta
- Department of Cell and Developmental Biology, Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gary C Hon
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jun Wu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA; Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
31
|
Huang H, Kuang X, Zou Y, Zeng J, Du H, Tang H, Long C, Mao Y, Yu X, Wen C, Yan J, Shen H. MAP4K4 is involved in the neuronal development of retinal photoreceptors. Exp Eye Res 2023; 233:109524. [PMID: 37290629 DOI: 10.1016/j.exer.2023.109524] [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: 02/14/2023] [Revised: 05/31/2023] [Accepted: 06/05/2023] [Indexed: 06/10/2023]
Abstract
Mitogen-activated protein kinase kinase kinase kinase-4 (MAP4K4) is a potential regulator of photoreceptor development. To investigate the mechanisms underlying MAP4K4 during the neuronal development of retinal photoreceptors, we generated knockout models of C57BL/6j mice in vivo and 661 W cells in vitro. Our findings revealed homozygous lethality and neural tube malformation in mice subjected to Map4k4 DNA ablation, providing evidence for the involvement of MAP4K4 in early stage embryonic neural formation. Furthermore, our study demonstrated that the ablation of Map4k4 DNA led to the vulnerability of photoreceptor neurites during induced neuronal development. By monitoring transcriptional and protein variations in mitogen-activated protein kinase (MAPK) signaling pathway-related factors, we discovered an imbalance in neurogenesis-related factors in Map4k4 -/- cells. Specifically, MAP4K4 promotes jun proto-oncogene (c-JUN) phosphorylation and recruits other factors related to nerve growth, ultimately leading to the robust formation of photoreceptor neurites. These data suggest that MAP4K4 plays a decisive role in regulating the fate of retinal photoreceptors through molecular modulation and contributes to our understanding of vision formation.
Collapse
Affiliation(s)
- Hao Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China; Department of Ophthalmology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, China
| | - Xielan Kuang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China; Biobank of Eye, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Yuxiu Zou
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Jingshu Zeng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Han Du
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Han Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Chongde Long
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Yan Mao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Xinyue Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Chaojuan Wen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Jianhua Yan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| | - Huangxuan Shen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China; Biobank of Eye, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China.
| |
Collapse
|
32
|
Adair BA, Korecki AJ, Djaksigulova D, Wagner PK, Chiu NY, Lam SL, Lengyell TC, Leavitt BR, Simpson EM. ABE8e Corrects Pax6-Aniridic Variant in Humanized Mouse ESCs and via LNPs in Ex Vivo Cortical Neurons. Ophthalmol Ther 2023; 12:2049-2068. [PMID: 37210469 PMCID: PMC10287867 DOI: 10.1007/s40123-023-00729-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 04/27/2023] [Indexed: 05/22/2023] Open
Abstract
INTRODUCTION Aniridia is a rare congenital vision-loss disease caused by heterozygous variants in the PAX6 gene. There is no vision-saving therapy, but one exciting approach is to use CRISPR/Cas9 to permanently correct the causal genomic variants. Preclinical studies to develop such a therapy in animal models face the challenge of showing efficacy when binding human DNA. Thus, we hypothesized that a CRISPR gene therapy can be developed and optimized in humanized mouse embryonic stem cells (ESCs) that will be able to distinguish between an aniridia patient variant and nonvariant chromosome and lay the foundation for human therapy. METHODS To answer the challenge of binding human DNA, we proposed the "CRISPR Humanized Minimally Mouse Models" (CHuMMMs) strategy. Thus, we minimally humanized Pax6 exon 9, the location of the most common aniridia variant c.718C > T. We generated and characterized a nonvariant CHuMMMs mouse, and a CHuMMMs cell-based disease model, in which we tested five CRISPR enzymes for therapeutic efficacy. We then delivered the therapy via lipid nanoparticles (LNPs) to alter a second variant in ex vivo cortical primary neurons. RESULTS We successfully established a nonvariant CHuMMMs mouse and three novel CHuMMMs aniridia cell lines. We showed that humanization did not disrupt Pax6 function in vivo, as the mouse showed no ocular phenotype. We developed and optimized a CRISPR therapeutic strategy for aniridia in the in vitro system, and found that the base editor, ABE8e, had the highest correction of the patient variant at 76.8%. In the ex vivo system, the LNP-encapsulated ABE8e ribonucleoprotein (RNP) complex altered the second patient variant and rescued 24.8% Pax6 protein expression. CONCLUSION We demonstrated the usefulness of the CHuMMMs approach, and showed the first genomic editing by ABE8e encapsulated as an LNP-RNP. Furthermore, we laid the foundation for translation of the proposed CRISPR therapy to preclinical mouse studies and eventually patients with aniridia.
Collapse
Affiliation(s)
- Bethany A Adair
- Centre for Molecular Medicine and Therapeutics at British Columbia Children's Hospital, The University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, The University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
| | - Andrea J Korecki
- Centre for Molecular Medicine and Therapeutics at British Columbia Children's Hospital, The University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
| | - Diana Djaksigulova
- Centre for Molecular Medicine and Therapeutics at British Columbia Children's Hospital, The University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
| | | | - Nina Y Chiu
- Centre for Molecular Medicine and Therapeutics at British Columbia Children's Hospital, The University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, The University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
| | - Siu Ling Lam
- Centre for Molecular Medicine and Therapeutics at British Columbia Children's Hospital, The University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
| | - Tess C Lengyell
- Centre for Molecular Medicine and Therapeutics at British Columbia Children's Hospital, The University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
| | - Blair R Leavitt
- Centre for Molecular Medicine and Therapeutics at British Columbia Children's Hospital, The University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- Department of Medical Genetics, The University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada
- Incisive Genetics Inc., Vancouver, BC, Canada
| | - Elizabeth M Simpson
- Centre for Molecular Medicine and Therapeutics at British Columbia Children's Hospital, The University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
- Department of Medical Genetics, The University of British Columbia, 950 West 28th Avenue, Vancouver, BC, V5Z 4H4, Canada.
| |
Collapse
|
33
|
Watson A, Lako M. Retinal organoids provide unique insights into molecular signatures of inherited retinal disease throughout retinogenesis. J Anat 2023; 243:186-203. [PMID: 36177499 PMCID: PMC10335378 DOI: 10.1111/joa.13768] [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: 06/17/2022] [Revised: 09/06/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022] Open
Abstract
The demand for induced pluripotent stem cells (iPSC)-derived retinal organoid and retinal pigment epithelium (RPE) models for the modelling of inherited retinopathies has increased significantly in the last decade. These models are comparable with foetal retinas up until the later stages of retinogenesis, expressing all of the key neuronal markers necessary for retinal function. These models have proven to be invaluable in the understanding of retinogenesis, particular in the context of patient-specific diseases. Inherited retinopathies are infamously described as clinically and phenotypically heterogeneous, such that developing gene/mutation-specific animal models in each instance of retinal disease is not financially or ethically feasible. Further to this, many animal models are insufficient in the study of disease pathogenesis due to anatomical differences and failure to recapitulate human disease phenotypes. In contrast, iPSC-derived retinal models provide a high throughput platform which is physiologically relevant for studying human health and disease. They also serve as a platform for drug screening, gene therapy approaches and in vitro toxicology of novel therapeutics in pre-clinical studies. One unique characteristic of stem cell-derived retinal models is the ability to mimic in vivo retinogenesis, providing unparalleled insights into the effects of pathogenic mutations in cells of the developing retina, in a highly accessible way. This review aims to give the reader an overview of iPSC-derived retinal organoids and/or RPE in the context of disease modelling of several inherited retinopathies including Retinitis Pigmentosa, Stargardt disease and Retinoblastoma. We describe the ability of each model to recapitulate in vivo disease phenotypes, validate previous findings from animal models and identify novel pathomechanisms that underpin individual IRDs.
Collapse
Affiliation(s)
- Avril Watson
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Majlinda Lako
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| |
Collapse
|
34
|
Kim JM, Min KW, Kim YJ, Smits R, Basler K, Kim JW. Wnt/β-Catenin Signaling Pathway Is Necessary for the Specification but Not the Maintenance of the Mouse Retinal Pigment Epithelium. Mol Cells 2023; 46:441-450. [PMID: 37190767 PMCID: PMC10336276 DOI: 10.14348/molcells.2023.0029] [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: 02/06/2023] [Accepted: 03/19/2023] [Indexed: 05/17/2023] Open
Abstract
β-Catenin (Ctnnb1) has been shown to play critical roles in the development and maintenance of epithelial cells, including the retinal pigment epithelium (RPE). Ctnnb1 is not only a component of intercellular junctions in the epithelium, it also functions as a transcriptional regulator in the Wnt signaling pathway. To identify which of its functional modalities is critically involved in mouse RPE development and maintenance, we varied Ctnnb1 gene content and activity in mouse RPE lineage cells and tested their impacts on mouse eye development. We found that a Ctnnb1 double mutant (Ctnnb1dm), which exhibits impaired transcriptional activity, could not replace Ctnnb1 in the RPE, whereas Ctnnb1Y654E, which has reduced affinity for the junctions, could do so. Expression of the constitutively active Ctnnb1∆ex3 mutant also suppressed the development of RPE, instead facilitating a ciliary cell fate. However, the post-mitotic or mature RPE was insensitive to the loss, inactivation, or constitutive activation of Ctnnb1. Collectively, our results suggest that Ctnnb1 should be maintained within an optimal range to specify RPE through transcriptional regulation of Wnt target genes in the optic neuroepithelium.
Collapse
Affiliation(s)
- Jong-Myeong Kim
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Kwang Wook Min
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - You-Joung Kim
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Ron Smits
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Konrad Basler
- Department of Molecular Life Sciences, University of Zurich, CH-8057 Zurich, Switzerland
| | - Jin Woo Kim
- Department of Biological Sciences and KAIST Stem Cell Center, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
35
|
Cullen PF, Sun D. Astrocytes of the eye and optic nerve: heterogeneous populations with unique functions mediate axonal resilience and vulnerability to glaucoma. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1217137. [PMID: 37829657 PMCID: PMC10569075 DOI: 10.3389/fopht.2023.1217137] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
The role of glia, particularly astrocytes, in mediating the central nervous system's response to injury and neurodegenerative disease is an increasingly well studied topic. These cells perform myriad support functions under physiological conditions but undergo behavioral changes - collectively referred to as 'reactivity' - in response to the disruption of neuronal homeostasis from insults, including glaucoma. However, much remains unknown about how reactivity alters disease progression - both beneficially and detrimentally - and whether these changes can be therapeutically modulated to improve outcomes. Historically, the heterogeneity of astrocyte behavior has been insufficiently addressed under both physiological and pathological conditions, resulting in a fragmented and often contradictory understanding of their contributions to health and disease. Thanks to increased focus in recent years, we now know this heterogeneity encompasses both intrinsic variation in physiological function and insult-specific changes that vary between pathologies. Although previous studies demonstrate astrocytic alterations in glaucoma, both in human disease and animal models, generally these findings do not conclusively link astrocytes to causative roles in neuroprotection or degeneration, rather than a subsequent response. Efforts to bolster our understanding by drawing on knowledge of brain astrocytes has been constrained by the primacy in the literature of findings from peri-synaptic 'gray matter' astrocytes, whereas much early degeneration in glaucoma occurs in axonal regions populated by fibrous 'white matter' astrocytes. However, by focusing on findings from astrocytes of the anterior visual pathway - those of the retina, unmyelinated optic nerve head, and myelinated optic nerve regions - we aim to highlight aspects of their behavior that may contribute to axonal vulnerability and glaucoma progression, including roles in mitochondrial turnover and energy provisioning. Furthermore, we posit that astrocytes of the retina, optic nerve head and myelinated optic nerve, although sharing developmental origins and linked by a network of gap junctions, may be best understood as distinct populations residing in markedly different niches with accompanying functional specializations. A closer investigation of their behavioral repertoires may elucidate not only their role in glaucoma, but also mechanisms to induce protective behaviors that can impede the progressive axonal damage and retinal ganglion cell death that drive vision loss in this devastating condition.
Collapse
Affiliation(s)
- Paul F. Cullen
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
| | - Daniel Sun
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
36
|
Luis J, Eastlake K, Lamb WDB, Limb GA, Jayaram H, Khaw PT. Cell-Based Therapies for Glaucoma. Transl Vis Sci Technol 2023; 12:23. [PMID: 37494052 PMCID: PMC10383000 DOI: 10.1167/tvst.12.7.23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 06/20/2023] [Indexed: 07/27/2023] Open
Abstract
Glaucomatous optic neuropathy (GON) is the major cause of irreversible visual loss worldwide and can result from a range of disease etiologies. The defining features of GON are retinal ganglion cell (RGC) degeneration and characteristic cupping of the optic nerve head (ONH) due to tissue remodeling, while intraocular pressure remains the only modifiable GON risk factor currently targeted by approved clinical treatment strategies. Efforts to understand the mechanisms that allow species such as the zebrafish to regenerate their retinal cells have greatly increased our understanding of regenerative signaling pathways. However, proper integration within the retina and projection to the brain by the newly regenerated neuronal cells remain major hurdles. Meanwhile, a range of methods for in vitro differentiation have been developed to derive retinal cells from a variety of cell sources, including embryonic and induced pluripotent stem cells. More recently, there has been growing interest in the implantation of glial cells as well as cell-derived products, including neurotrophins, microRNA, and extracellular vesicles, to provide functional support to vulnerable structures such as RGC axons and the ONH. These approaches offer the advantage of not relying upon the replacement of degenerated cells and potentially targeting earlier stages of disease pathogenesis. In order to translate these techniques into clinical practice, appropriate cell sourcing, robust differentiation protocols, and accurate implantation methods are crucial to the success of cell-based therapy in glaucoma. Translational Relevance: Cell-based therapies for glaucoma currently under active development include the induction of endogenous regeneration, implantation of exogenously derived retinal cells, and utilization of cell-derived products to provide functional support.
Collapse
Affiliation(s)
- Joshua Luis
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - Karen Eastlake
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - William D. B. Lamb
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - G. Astrid Limb
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - Hari Jayaram
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| | - Peng T. Khaw
- NIHR Biomedical Research Centre for Ophthalmology, UCL Institute of Ophthalmology & Moorfields Eye Hospital, London, UK
| |
Collapse
|
37
|
Fanaro GB, Marques MR, Calaza KDC, Brito R, Pessoni AM, Mendonça HR, Lemos DEDA, de Brito Alves JL, de Souza EL, Cavalcanti Neto MP. New Insights on Dietary Polyphenols for the Management of Oxidative Stress and Neuroinflammation in Diabetic Retinopathy. Antioxidants (Basel) 2023; 12:1237. [PMID: 37371967 PMCID: PMC10295526 DOI: 10.3390/antiox12061237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Diabetic retinopathy (DR) is a neurodegenerative and vascular pathology that is considered one of the leading causes of blindness worldwide, resulting from complications of advanced diabetes mellitus (DM). Current therapies consist of protocols aiming to alleviate the existing clinical signs associated with microvascular alterations limited to the advanced disease stages. In response to the low resolution and limitations of the DR treatment, there is an urgent need to develop more effective alternative therapies to optimize glycemic, vascular, and neuronal parameters, including the reduction in the cellular damage promoted by inflammation and oxidative stress. Recent evidence has shown that dietary polyphenols reduce oxidative and inflammatory parameters of various diseases by modulating multiple cell signaling pathways and gene expression, contributing to the improvement of several chronic diseases, including metabolic and neurodegenerative diseases. However, despite the growing evidence for the bioactivities of phenolic compounds, there is still a lack of data, especially from human studies, on the therapeutic potential of these substances. This review aims to comprehensively describe and clarify the effects of dietary phenolic compounds on the pathophysiological mechanisms involved in DR, especially those of oxidative and inflammatory nature, through evidence from experimental studies. Finally, the review highlights the potential of dietary phenolic compounds as a prophylactic and therapeutic strategy and the need for further clinical studies approaching the efficacy of these substances in DR management.
Collapse
Affiliation(s)
- Gustavo Bernardes Fanaro
- Institute of Health and Biotechnology, Federal University of Amazonas, Manaus 69460000, Amazonas, Brazil;
| | | | - Karin da Costa Calaza
- Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói 24210201, Rio de Janeiro, Brazil;
| | - Rafael Brito
- Department of Cellular and Molecular Biology, Institute of Biology, Fluminense Federal University, Niterói 24210201, Rio de Janeiro, Brazil;
| | | | - Henrique Rocha Mendonça
- Institute of Biodiversity and Sustainability (NUPEM), Federal University of Rio de Janeiro, Macaé 27965045, Rio de Janeiro, Brazil; (H.R.M.); (M.P.C.N.)
| | | | - José Luiz de Brito Alves
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, João Pessoa 58051900, Paraíba, Brazil; (D.E.d.A.L.); (J.L.d.B.A.)
| | - Evandro Leite de Souza
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, João Pessoa 58051900, Paraíba, Brazil; (D.E.d.A.L.); (J.L.d.B.A.)
| | - Marinaldo Pacífico Cavalcanti Neto
- Institute of Biodiversity and Sustainability (NUPEM), Federal University of Rio de Janeiro, Macaé 27965045, Rio de Janeiro, Brazil; (H.R.M.); (M.P.C.N.)
| |
Collapse
|
38
|
Li R, Liu J, Yi P, Yang X, Chen J, Zhao C, Liao X, Wang X, Xu Z, Lu H, Li H, Zhang Z, Liu X, Xiang J, Hu K, Qi H, Yu J, Yang P, Hou S. Integrative Single-Cell Transcriptomics and Epigenomics Mapping of the Fetal Retina Developmental Dynamics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206623. [PMID: 37017569 DOI: 10.1002/advs.202206623] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 02/24/2023] [Indexed: 06/04/2023]
Abstract
The underlying mechanisms that determine gene expression and chromatin accessibility in retinogenesis are poorly understood. Herein, single-cell RNA sequencing and single-cell assay for transposase-accessible chromatin sequencing are performed on human embryonic eye samples obtained 9-26 weeks after conception to explore the heterogeneity of retinal progenitor cells (RPCs) and neurogenic RPCs. The differentiation trajectory from RPCs to 7 major types of retinal cells are verified. Subsequently, diverse lineage-determining transcription factors are identified and their gene regulatory networks are refined at the transcriptomic and epigenomic levels. Treatment of retinospheres, with the inhibitor of RE1 silencing transcription factor, X5050, induces more neurogenesis with the regular arrangement, and a decrease in Müller glial cells. The signatures of major retinal cells and their correlation with pathogenic genes associated with multiple ocular diseases, including uveitis and age-related macular degeneration are also described. A framework for the integrated exploration of single-cell developmental dynamics of the human primary retina is provided.
Collapse
Affiliation(s)
- Ruonan Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Jiangyi Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Ping Yi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, P. R. China
| | - Xianli Yang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, P. R. China
| | - Jun Chen
- Department of Obstetrics, Women and Children's Hospital of Chongqing Medical University, Chongqing, 401147, P. R. China
| | - Chenyang Zhao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Xingyun Liao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, P. R. China
| | - Xiaotang Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Zongren Xu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
| | - Huiping Lu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Hongshun Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Zhi Zhang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Xianyang Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Junjie Xiang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Ke Hu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Hongbo Qi
- Department of Obstetrics, Women and Children's Hospital of Chongqing Medical University, Chongqing, 401147, P. R. China
- Chongqing Key Laboratory of Maternal and Fetal Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Jia Yu
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Haihe Laboratory of Cell Ecosystem, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, 100005, P. R. China
- The Key Laboratory of RNA and Hematopoietic Regulation, Chinese Academy of Medical Sciences, Beijing, 100005, P. R. China
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
| | - Shengping Hou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
- Chongqing Key Laboratory of Ophthalmology, Chongqing, 400016, P. R. China
- Chongqing Eye Institute, Chongqing, 400016, P. R. China
- Chongqing Branch (Municipality Division) of National Clinical Research Center for Ocular Diseases, Chongqing, 400016, P. R. China
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Beijing Ophthalmology & Visual Sciences Key Laboratory, Beijing, 100730, P. R. China
| |
Collapse
|
39
|
Paşcalău R, Badea TC. Signaling - transcription interactions in mouse retinal ganglion cells early axon pathfinding -a literature review. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1180142. [PMID: 38983012 PMCID: PMC11182120 DOI: 10.3389/fopht.2023.1180142] [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: 03/05/2023] [Accepted: 04/21/2023] [Indexed: 07/11/2024]
Abstract
Sending an axon out of the eye and into the target brain nuclei is the defining feature of retinal ganglion cells (RGCs). The literature on RGC axon pathfinding is vast, but it focuses mostly on decision making events such as midline crossing at the optic chiasm or retinotopic mapping at the target nuclei. In comparison, the exit of RGC axons out of the eye is much less explored. The first checkpoint on the RGC axons' path is the optic cup - optic stalk junction (OC-OS). OC-OS development and the exit of the RGC pioneer axons out of the eye are coordinated spatially and temporally. By the time the optic nerve head domain is specified, the optic fissure margins are in contact and the fusion process is ongoing, the first RGCs are born in its proximity and send pioneer axons in the optic stalk. RGC differentiation continues in centrifugal waves. Later born RGC axons fasciculate with the more mature axons. Growth cones at the end of the axons respond to guidance cues to adopt a centripetal direction, maintain nerve fiber layer restriction and to leave the optic cup. Although there is extensive information on OC-OS development, we still have important unanswered questions regarding its contribution to the exit of the RGC axons out of the eye. We are still to distinguish the morphogens of the OC-OS from the axon guidance molecules which are expressed in the same place at the same time. The early RGC transcription programs responsible for axon emergence and pathfinding are also unknown. This review summarizes the molecular mechanisms for early RGC axon guidance by contextualizing mouse knock-out studies on OC-OS development with the recent transcriptomic studies on developing RGCs in an attempt to contribute to the understanding of human optic nerve developmental anomalies. The published data summarized here suggests that the developing optic nerve head provides a physical channel (the closing optic fissure) as well as molecular guidance cues for the pioneer RGC axons to exit the eye.
Collapse
Affiliation(s)
- Raluca Paşcalău
- Research and Development Institute, Transilvania University of Braşov, Braşov, Romania
- Ophthalmology Clinic, Cluj County Emergency Hospital, Cluj-Napoca, Romania
| | - Tudor Constantin Badea
- Research and Development Institute, Transilvania University of Braşov, Braşov, Romania
- National Center for Brain Research, Institutul de Cercetări pentru Inteligență Artificială, Romanian Academy, Bucharest, Romania
| |
Collapse
|
40
|
Yu D, Wu Y, Zhu L, Wang Y, Sheng D, Zhao X, Liang G, Gan L. The landscape of the long non-coding RNAs in developing mouse retinas. BMC Genomics 2023; 24:252. [PMID: 37165305 PMCID: PMC10173636 DOI: 10.1186/s12864-023-09354-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 05/03/2023] [Indexed: 05/12/2023] Open
Abstract
BACKGROUND The long non-coding RNAs (lncRNAs) are critical regulators of diverse biological processes. Nevertheless, a global view of its expression and function in the mouse retina, a crucial model for neurogenesis study, still needs to be made available. RESULTS Herein, by integrating the established gene models and the result from ab initio prediction using short- and long-read sequencing, we characterized 4,523 lncRNA genes (MRLGs) in developing mouse retinas (from the embryonic day of 12.5 to the neonatal day of P28), which was so far the most comprehensive collection of retinal lncRNAs. Next, derived from transcriptomics analyses of different tissues and developing retinas, we found that the MRLGs were highly spatiotemporal specific in expression and played essential roles in regulating the genesis and function of mouse retinas. In addition, we investigated the expression of MRLGs in some mouse mutants and revealed that 97 intergenic MRLGs might be involved in regulating differentiation and development of retinal neurons through Math5, Isl1, Brn3b, NRL, Onecut1, or Onecut2 mediated pathways. CONCLUSIONS In summary, this work significantly enhanced our knowledge of lncRNA genes in mouse retina development and provided valuable clues for future exploration of their biological roles.
Collapse
Affiliation(s)
- Dongliang Yu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, 310018, China.
- Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China.
| | - Yuqing Wu
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, 310018, China
| | - Leilei Zhu
- Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Yuying Wang
- Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Donglai Sheng
- Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Xiaofeng Zhao
- Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China
| | - Guoqing Liang
- Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang, 310036, China.
| | - Lin Gan
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
41
|
Yu J, Yin Y, Leng Y, Zhang J, Wang C, Chen Y, Li X, Wang X, Liu H, Liao Y, Jin Y, Zhang Y, Lu K, Wang K, Wang X, Wang L, Zheng F, Gu Z, Li Y, Fan Y. Emerging strategies of engineering retinal organoids and organoid-on-a-chip in modeling intraocular drug delivery: current progress and future perspectives. Adv Drug Deliv Rev 2023; 197:114842. [PMID: 37105398 DOI: 10.1016/j.addr.2023.114842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/17/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023]
Abstract
Retinal diseases are a rising concern as major causes of blindness in an aging society; therapeutic options are limited, and the precise pathogenesis of these diseases remains largely unknown. Intraocular drug delivery and nanomedicines offering targeted, sustained, and controllable delivery are the most challenging and popular topics in ocular drug development and toxicological evaluation. Retinal organoids (ROs) and organoid-on-a-chip (ROoC) are both emerging as promising in-vitro models to faithfully recapitulate human eyes for retinal research in the replacement of experimental animals and primary cells. In this study, we review the generation and application of ROs resembling the human retina in cell subtypes and laminated structures and introduce the emerging engineered ROoC as a technological opportunity to address critical issues. On-chip vascularization, perfusion, and close inter-tissue interactions recreate physiological environments in vitro, whilst integrating with biosensors facilitates real-time analysis and monitoring during organogenesis of the retina representing engineering efforts in ROoC models. We also emphasize that ROs and ROoCs hold the potential for applications in modeling intraocular drug delivery in vitro and developing next-generation retinal drug delivery strategies.
Collapse
Affiliation(s)
- Jiaheng Yu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yuqi Yin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yubing Leng
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Jingcheng Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Chunyan Wang
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China
| | - Yanyun Chen
- Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Xiaorui Li
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Xudong Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Hui Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yulong Liao
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yishan Jin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Yihan Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Keyu Lu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Kehao Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China
| | - Xiaofei Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China
| | - Lizhen Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China
| | - Fuyin Zheng
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China.
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Yinghui Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, 100094, China.
| | - Yubo Fan
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, and with the School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China; Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing, 100083, China.
| |
Collapse
|
42
|
Chinnaiya K, Burbridge S, Jones A, Kim DW, Place E, Manning E, Groves I, Sun C, Towers M, Blackshaw S, Placzek M. A neuroepithelial wave of BMP signalling drives anteroposterior specification of the tuberal hypothalamus. eLife 2023; 12:e83133. [PMID: 36718990 PMCID: PMC9917434 DOI: 10.7554/elife.83133] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 01/29/2023] [Indexed: 02/01/2023] Open
Abstract
The tuberal hypothalamus controls life-supporting homeostatic processes, but despite its fundamental role, the cells and signalling pathways that specify this unique region of the central nervous system in embryogenesis are poorly characterised. Here, we combine experimental and bioinformatic approaches in the embryonic chick to show that the tuberal hypothalamus is progressively generated from hypothalamic floor plate-like cells. Fate-mapping studies show that a stream of tuberal progenitors develops in the anterior-ventral neural tube as a wave of neuroepithelial-derived BMP signalling sweeps from anterior to posterior through the hypothalamic floor plate. As later-specified posterior tuberal progenitors are generated, early specified anterior tuberal progenitors become progressively more distant from these BMP signals and differentiate into tuberal neurogenic cells. Gain- and loss-of-function experiments in vivo and ex vivo show that BMP signalling initiates tuberal progenitor specification, but must be eliminated for these to progress to anterior neurogenic progenitors. scRNA-Seq profiling shows that tuberal progenitors that are specified after the major period of anterior tuberal specification begin to upregulate genes that characterise radial glial cells. This study provides an integrated account of the development of the tuberal hypothalamus.
Collapse
Affiliation(s)
- Kavitha Chinnaiya
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Sarah Burbridge
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Aragorn Jones
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Dong Won Kim
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Elsie Place
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Elizabeth Manning
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Ian Groves
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Changyu Sun
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Matthew Towers
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Psychiatry and Behavioral Science, 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
| | - Marysia Placzek
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
- Bateson Centre, University of SheffieldSheffieldUnited Kingdom
- Neuroscience Institute, University of SheffieldSheffieldUnited Kingdom
| |
Collapse
|
43
|
Chee JM, Lanoue L, Clary D, Higgins K, Bower L, Flenniken A, Guo R, Adams DJ, Bosch F, Braun RE, Brown SDM, Chin HJG, Dickinson ME, Hsu CW, Dobbie M, Gao X, Galande S, Grobler A, Heaney JD, Herault Y, de Angelis MH, Mammano F, Nutter LMJ, Parkinson H, Qin C, Shiroishi T, Sedlacek R, Seong JK, Xu Y, Brooks B, McKerlie C, Lloyd KCK, Westerberg H, Moshiri A. Genome-wide screening reveals the genetic basis of mammalian embryonic eye development. BMC Biol 2023; 21:22. [PMID: 36737727 PMCID: PMC9898963 DOI: 10.1186/s12915-022-01475-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 11/23/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Microphthalmia, anophthalmia, and coloboma (MAC) spectrum disease encompasses a group of eye malformations which play a role in childhood visual impairment. Although the predominant cause of eye malformations is known to be heritable in nature, with 80% of cases displaying loss-of-function mutations in the ocular developmental genes OTX2 or SOX2, the genetic abnormalities underlying the remaining cases of MAC are incompletely understood. This study intended to identify the novel genes and pathways required for early eye development. Additionally, pathways involved in eye formation during embryogenesis are also incompletely understood. This study aims to identify the novel genes and pathways required for early eye development through systematic forward screening of the mammalian genome. RESULTS Query of the International Mouse Phenotyping Consortium (IMPC) database (data release 17.0, August 01, 2022) identified 74 unique knockout lines (genes) with genetically associated eye defects in mouse embryos. The vast majority of eye abnormalities were small or absent eyes, findings most relevant to MAC spectrum disease in humans. A literature search showed that 27 of the 74 lines had previously published knockout mouse models, of which only 15 had ocular defects identified in the original publications. These 12 previously published gene knockouts with no reported ocular abnormalities and the 47 unpublished knockouts with ocular abnormalities identified by the IMPC represent 59 genes not previously associated with early eye development in mice. Of these 59, we identified 19 genes with a reported human eye phenotype. Overall, mining of the IMPC data yielded 40 previously unimplicated genes linked to mammalian eye development. Bioinformatic analysis showed that several of the IMPC genes colocalized to several protein anabolic and pluripotency pathways in early eye development. Of note, our analysis suggests that the serine-glycine pathway producing glycine, a mitochondrial one-carbon donator to folate one-carbon metabolism (FOCM), is essential for eye formation. CONCLUSIONS Using genome-wide phenotype screening of single-gene knockout mouse lines, STRING analysis, and bioinformatic methods, this study identified genes heretofore unassociated with MAC phenotypes providing models to research novel molecular and cellular mechanisms involved in eye development. These findings have the potential to hasten the diagnosis and treatment of this congenital blinding disease.
Collapse
Affiliation(s)
- Justine M Chee
- Oakland University William Beaumont School of Medicine, Rochester, MI, USA
| | - Louise Lanoue
- Mouse Biology Program, University of California Davis, Davis, CA, USA
| | - Dave Clary
- Mouse Biology Program, University of California Davis, Davis, CA, USA
| | - Kendall Higgins
- University of Miami: Miller School of Medicine, Miami, FL, USA
| | - Lynette Bower
- Mouse Biology Program, University of California Davis, Davis, CA, USA
| | - Ann Flenniken
- The Centre for Phenogenomics, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, ON, Canada
| | - Ruolin Guo
- The Centre for Phenogenomics, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | - David J Adams
- The Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Fatima Bosch
- Centre of Animal Biotechnology and Gene Therapy (CBATEG), Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Steve D M Brown
- Medical Research Council Harwell Institute, Mammalian Genetics Unit and Mary Lyon Centre, Harwell Campus, Oxfordshire, UK
| | - H-J Genie Chin
- National Laboratory Animal Center, National Applied Research Laboratories (NARLabs), Taipei City, Taiwan
| | - Mary E Dickinson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Chih-Wei Hsu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Michael Dobbie
- Phenomics Australia, The John Curtin School of Medical Research, Canberra, Australia
| | - Xiang Gao
- Nanjing Biomedical Research Institute, Nanjing University, Nanjing, China
| | - Sanjeev Galande
- Indian Institutes of Science Education and Research, Pune, India
| | - Anne Grobler
- Faculty of Health Sciences, PCDDP North-West University, Potchefstroom, South Africa
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Yann Herault
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, Illkirch, France
| | - Martin Hrabe de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Fabio Mammano
- Monterotondo Mouse Clinic, Italian National Research Council (CNR), Monterotondo Scalo, Italy
| | - Lauryl M J Nutter
- The Centre for Phenogenomics, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | - Helen Parkinson
- European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridgeshire, UK
| | - Chuan Qin
- National Laboratory Animal Center, National Applied Research Laboratories, Beijing, China
| | | | - Radislav Sedlacek
- Czech Center for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic
| | - J-K Seong
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea
| | - Ying Xu
- CAM-SU Genomic Resource Center, Soochow University, Suzhou, China
| | - Brian Brooks
- Ophthalmic Genetics and Visual Function Branch, National Eye Institute, NIH, Bethesda, MD, 20892, USA
| | - Colin McKerlie
- The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine & Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - K C Kent Lloyd
- Mouse Biology Program, University of California Davis, Davis, CA, USA
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Henrik Westerberg
- Medical Research Council Harwell Institute, Mammalian Genetics Unit and Mary Lyon Centre, Harwell Campus, Oxfordshire, UK
| | - Ala Moshiri
- Department of Ophthalmology & Vision Science, School of Medicine, University of California Davis, Sacramento, CA, USA.
- UC Davis Eye Center, 4860 Y St., Ste. 2400, Sacramento, CA, 95817, USA.
| |
Collapse
|
44
|
Blommers M, Stanton-Turcotte D, Iulianella A. Retinal neuroblast migration and ganglion cell layer organization require the cytoskeletal-interacting protein Mllt11. Dev Dyn 2023; 252:305-319. [PMID: 36131367 DOI: 10.1002/dvdy.540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 09/16/2022] [Accepted: 09/18/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The vertebrate retina is an organized laminar structure comprised of distinct cell types populating three nuclear layers. During development, each retinal cell type follows a stereotypical temporal order of genesis, differentiation, and migration, giving rise to its stratified organization. Once born, the precise positioning of cells along the apico-basal (radial) axis of the retina is critical for subsequent connections to form, relying on highly orchestrated migratory processes. While these processes are critical for visual function to arise, the regulators of cellular migration and retinal lamination remain largely unexplored. RESULTS We report a role for a microtubule-interacting protein, Mllt11 (myeloid/lymphoid or mixed-lineage leukemia; translocated to chromosome 11/All1 fused gene from chromosome 1q) in mammalian retinal cell migration during retinogenesis. We show that Mllt11 loss-of-function in mouse retinal neuroblasts affected the migration of ganglion and amacrine cells into the ganglion cell layer and led to their aberrant accumulation in the inner nuclear and plexiform layers. CONCLUSIONS We demonstrate a role for Mllt11 in neuroblast migration and formation of the ganglion cell layer of the retina.
Collapse
Affiliation(s)
- Marley Blommers
- Department of Medical Neuroscience, and Brain Repair Centre, Faculty of Medicine, Life Science Research Institute, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Danielle Stanton-Turcotte
- Department of Medical Neuroscience, and Brain Repair Centre, Faculty of Medicine, Life Science Research Institute, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Angelo Iulianella
- Department of Medical Neuroscience, and Brain Repair Centre, Faculty of Medicine, Life Science Research Institute, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
45
|
Future regenerative medicine developments and their therapeutic applications. Biomed Pharmacother 2023; 158:114131. [PMID: 36538861 DOI: 10.1016/j.biopha.2022.114131] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Although the currently available pharmacological assays can cure most pathological disorders, they have limited therapeutic value in relieving certain disorders like myocardial infarct, peripheral vascular disease, amputated limbs, or organ failure (e.g. renal failure). Pilot studies to overcome such problems using regenerative medicine (RM) delivered promising data. Comprehensive investigations of RM in zebrafish or reptilians are necessary for better understanding. However, the precise mechanisms remain poorly understood despite the tremendous amount of data obtained using the zebrafish model investigating the exact mechanisms behind their regenerative capability. Indeed, understanding such mechanisms and their application to humans can save millions of lives from dying due to potentially life-threatening events. Recent studies have launched a revolution in replacing damaged human organs via different approaches in the last few decades. The newly established branch of medicine (known as Regenerative Medicine aims to enhance natural repair mechanisms. This can be done through the application of several advanced broad-spectrum technologies such as organ transplantation, tissue engineering, and application of Scaffolds technology (support vascularization using an extracellular matrix), stem cell therapy, miRNA treatment, development of 3D mini-organs (organoids), and the construction of artificial tissues using nanomedicine and 3D bio-printers. Moreover, in the next few decades, revolutionary approaches in regenerative medicine will be applied based on artificial intelligence and wireless data exchange, soft intelligence biomaterials, nanorobotics, and even living robotics capable of self-repair. The present work presents a comprehensive overview that summarizes the new and future advances in the field of RM.
Collapse
|
46
|
Ziermann JM. Overview of Head Muscles with Special Emphasis on Extraocular Muscle Development. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2023; 236:57-80. [PMID: 37955771 DOI: 10.1007/978-3-031-38215-4_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
The head is often considered the most complex part of the vertebrate body as many different cell types contribute to a huge variation of structures in a very limited space. Most of these cell types also interact with each other to ensure the proper development of skull, brain, muscles, nerves, connective tissue, and blood vessels. While there are general mechanisms that are true for muscle development all over the body, the head and postcranial muscle development differ from each other. In the head, specific gene regulatory networks underlie the differentiation in subgroups, which include extraocular muscles, muscles of mastication, muscles of facial expression, laryngeal and pharyngeal muscles, as well as cranial nerve innervated neck muscles. Here, I provide an overview of the difference between head and trunk muscle development. This is followed by a short excursion to the cardiopharyngeal field which gives rise to heart and head musculature and a summary of pharyngeal arch muscle development, including interactions between neural crest cells, mesodermal cells, and endodermal signals. Lastly, a more detailed description of the eye development, tissue interactions, and involved genes is provided.
Collapse
|
47
|
Sun C, Chen S. Disease-causing mutations in genes encoding transcription factors critical for photoreceptor development. Front Mol Neurosci 2023; 16:1134839. [PMID: 37181651 PMCID: PMC10172487 DOI: 10.3389/fnmol.2023.1134839] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 04/04/2023] [Indexed: 05/16/2023] Open
Abstract
Photoreceptor development of the vertebrate visual system is controlled by a complex transcription regulatory network. OTX2 is expressed in the mitotic retinal progenitor cells (RPCs) and controls photoreceptor genesis. CRX that is activated by OTX2 is expressed in photoreceptor precursors after cell cycle exit. NEUROD1 is also present in photoreceptor precursors that are ready to specify into rod and cone photoreceptor subtypes. NRL is required for the rod fate and regulates downstream rod-specific genes including the orphan nuclear receptor NR2E3 which further activates rod-specific genes and simultaneously represses cone-specific genes. Cone subtype specification is also regulated by the interplay of several transcription factors such as THRB and RXRG. Mutations in these key transcription factors are responsible for ocular defects at birth such as microphthalmia and inherited photoreceptor diseases such as Leber congenital amaurosis (LCA), retinitis pigmentosa (RP) and allied dystrophies. In particular, many mutations are inherited in an autosomal dominant fashion, including the majority of missense mutations in CRX and NRL. In this review, we describe the spectrum of photoreceptor defects that are associated with mutations in the above-mentioned transcription factors, and summarize the current knowledge of molecular mechanisms underlying the pathogenic mutations. At last, we deliberate the outstanding gaps in our understanding of the genotype-phenotype correlations and outline avenues for future research of the treatment strategies.
Collapse
Affiliation(s)
- Chi Sun
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, United States
- *Correspondence: Chi Sun,
| | - Shiming Chen
- Department of Ophthalmology and Visual Sciences, Washington University in St. Louis, St. Louis, MO, United States
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO, United States
| |
Collapse
|
48
|
Lešin Gaćina D, Jandroković S, Vidas Pauk S, Pupić-Bakrač A, Vukojević N, Petriček I, Škegro I, Galiot Delić M. BILATERAL SEROUS CHORIORETINOPATHY AND PIGMENTARY GLAUCOMA - WHAT IS THE ASSOCIATION? Acta Clin Croat 2022; 61:727-734. [PMID: 37868183 PMCID: PMC10588375 DOI: 10.20471/acc.2022.61.04.22] [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: 06/24/2022] [Accepted: 11/03/2022] [Indexed: 10/24/2023] Open
Abstract
We present a patient with concurrent pigmentary glaucoma, bilateral central serous chorioretinopathy and unilateral optic disc pit, and propose a possible association of these conditions. Comprehensive ophthalmic examination of a 36-year-old man who was complaining of blurry vision and pain in the eyes showed reduced visual acuity on the left eye, elevated intraocular pressure in the right eye, bilateral signs of pigment dispersion syndrome, and bilateral central serous chorioretinopathy, combined with optic disc pit in the left eye. Visual field and optical coherence tomography findings demonstrated functional and structural glaucoma changes. Choroidal circulation abnormalities were observed by angiographic methods. Genetic and developmental anomalies of the external layer of the optic disc cup that gives rise to many anterior and posterior eye segment structures suggest a possible association of a clinical condition characterized by the combination of pigmentary glaucoma, central serous chorioretinopathy and optic disc pit. Future research would enable to determine proper diagnostic protocols, treatment and follow-up procedures for this chronic-progressive disorder.
Collapse
Affiliation(s)
- Dina Lešin Gaćina
- Department of Ophthalmology, Zagreb University Hospital Center, Zagreb, Croatia
| | - Sonja Jandroković
- Department of Ophthalmology, Zagreb University Hospital Center, Zagreb, Croatia
- University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Sania Vidas Pauk
- Department of Ophthalmology, Zagreb University Hospital Center, Zagreb, Croatia
| | - Ana Pupić-Bakrač
- Department of Ophthalmology, Zadar General Hospital, Zadar, Croatia
| | - Nenad Vukojević
- Department of Ophthalmology, Zagreb University Hospital Center, Zagreb, Croatia
- University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Igor Petriček
- Department of Ophthalmology, Zagreb University Hospital Center, Zagreb, Croatia
- University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Ivan Škegro
- Department of Ophthalmology, Zagreb University Hospital Center, Zagreb, Croatia
- University of Zagreb, School of Medicine, Zagreb, Croatia
| | | |
Collapse
|
49
|
Burgoyne T, Toms M, Way C, Tracey-White D, Futter CE, Moosajee M. Changes in Mitochondrial Size and Morphology in the RPE and Photoreceptors of the Developing and Ageing Zebrafish. Cells 2022; 11:cells11223542. [PMID: 36428971 PMCID: PMC9688747 DOI: 10.3390/cells11223542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/05/2022] [Accepted: 11/04/2022] [Indexed: 11/12/2022] Open
Abstract
Mitochondria are essential adenosine triphosphate (ATP)-generating cellular organelles. In the retina, they are highly numerous in the photoreceptors and retinal pigment epithelium (RPE) due to their high energetic requirements. Fission and fusion of the mitochondria within these cells allow them to adapt to changing demands over the lifespan of the organism. Using transmission electron microscopy, we examined the mitochondrial ultrastructure of zebrafish photoreceptors and RPE from 5 days post fertilisation (dpf) through to late adulthood (3 years). Notably, mitochondria in the youngest animals were large and irregular shaped with a loose cristae architecture, but by 8 dpf they had reduced in size and expanded in number with more defined cristae. Investigation of temporal gene expression of several mitochondrial-related markers indicated fission as the dominant mechanism contributing to the changes observed over time. This is likely to be due to continued mitochondrial stress resulting from the oxidative environment of the retina and prolonged light exposure. We have characterised retinal mitochondrial ageing in a key vertebrate model organism, that provides a basis for future studies of retinal diseases that are linked to mitochondrial dysfunction.
Collapse
Affiliation(s)
- Thomas Burgoyne
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK
- Royal Brompton Hospital, Guy’s and St Thomas’ NHS Foundation Trust, London SW3 6NP, UK
| | - Maria Toms
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK
- The Francis Crick Institute, London NW1 1AT, UK
| | - Chris Way
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Dhani Tracey-White
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Clare E. Futter
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK
| | - Mariya Moosajee
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, UK
- The Francis Crick Institute, London NW1 1AT, UK
- Department of Ophthalmology, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
- Department of Genetics, Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK
- Correspondence:
| |
Collapse
|
50
|
Transplanted human induced pluripotent stem cells- derived retinal ganglion cells embed within mouse retinas and are electrophysiologically functional. iScience 2022; 25:105308. [DOI: 10.1016/j.isci.2022.105308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/22/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022] Open
|