1
|
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
|
2
|
Zuo Z, Cheng X, Ferdous S, Shao J, Li J, Bao Y, Li J, Lu J, Jacobo Lopez A, Wohlschlegel J, Prieve A, Thomas MG, Reh TA, Li Y, Moshiri A, Chen R. Single cell dual-omic atlas of the human developing retina. Nat Commun 2024; 15:6792. [PMID: 39117640 PMCID: PMC11310509 DOI: 10.1038/s41467-024-50853-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
The development of the retina is under tight temporal and spatial control. To gain insights into the molecular basis of this process, we generate a single-nuclei dual-omic atlas of the human developing retina with approximately 220,000 nuclei from 14 human embryos and fetuses aged between 8 and 23-weeks post-conception with matched macular and peripheral tissues. This atlas captures all major cell classes in the retina, along with a large proportion of progenitors and cell-type-specific precursors. Cell trajectory analysis reveals a transition from continuous progression in early progenitors to a hierarchical development during the later stages of cell type specification. Both known and unrecorded candidate transcription factors, along with gene regulatory networks that drive the transitions of various cell fates, are identified. Comparisons between the macular and peripheral retinae indicate a largely consistent yet distinct developmental pattern. This atlas offers unparalleled resolution into the transcriptional and chromatin accessibility landscapes during development, providing an invaluable resource for deeper insights into retinal development and associated diseases.
Collapse
Affiliation(s)
- Zhen Zuo
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Xuesen Cheng
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Salma Ferdous
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Jianming Shao
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Jin Li
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Yourong Bao
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Jean Li
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Jiaxiong Lu
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Antonio Jacobo Lopez
- Department of Ophthalmology & Vision Science, UC Davis School of Medicine, 4860 Y St, Sacramento, CA, USA
| | - Juliette Wohlschlegel
- Department of Biological Structure, University of Washington, 1410 NE Campus Pkwy, Seattle, WA, USA
| | - Aric Prieve
- Department of Biological Structure, University of Washington, 1410 NE Campus Pkwy, Seattle, WA, USA
| | - Mervyn G Thomas
- Ulverscroft Eye Unit, School of Psychology and Vision Sciences, The University of Leicester, Leicester, UK
| | - Thomas A Reh
- Department of Biological Structure, University of Washington, 1410 NE Campus Pkwy, Seattle, WA, USA
| | - Yumei Li
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA
| | - Ala Moshiri
- Department of Ophthalmology & Vision Science, UC Davis School of Medicine, 4860 Y St, Sacramento, CA, USA
| | - Rui Chen
- HGSC, Department of Molecular and Human Genetics, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA.
- Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA.
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, USA.
- Gavin Herbert Eye Institute - Center for Translational Vision Research, Department of Ophthalmology, University of California Irvine School of Medicine, Irvine, USA.
| |
Collapse
|
3
|
Liu X, Pacwa A, Bresciani G, Swierczynska M, Dorecka M, Smedowski A. Retinal primary cilia and their dysfunction in retinal neurodegenerative diseases: beyond ciliopathies. Mol Med 2024; 30:109. [PMID: 39060957 PMCID: PMC11282803 DOI: 10.1186/s10020-024-00875-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Primary cilia are sensory organelles that extend from the cellular membrane and are found in a wide range of cell types. Cilia possess a plethora of vital components that enable the detection and transmission of several signaling pathways, including Wnt and Shh. In turn, the regulation of ciliogenesis and cilium length is influenced by various factors, including autophagy, organization of the actin cytoskeleton, and signaling inside the cilium. Irregularities in the development, maintenance, and function of this cellular component lead to a range of clinical manifestations known as ciliopathies. The majority of people with ciliopathies have a high prevalence of retinal degeneration. The most common theory is that retinal degeneration is primarily caused by functional and developmental problems within retinal photoreceptors. The contribution of other ciliated retinal cell types to retinal degeneration has not been explored to date. In this review, we examine the occurrence of primary cilia in various retinal cell types and their significance in pathology. Additionally, we explore potential therapeutic approaches targeting ciliopathies. By engaging in this endeavor, we present new ideas that elucidate innovative concepts for the future investigation and treatment of retinal ciliopathies.
Collapse
Affiliation(s)
- Xiaonan Liu
- Department of Ophthalmology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Ceglana 35, 40-514, Katowice, Poland.
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland.
| | - Anna Pacwa
- GlaucoTech Co, Katowice, Poland
- Department of Physiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Medykow 18, 40-752, Katowice, Poland
| | | | - Marta Swierczynska
- Department of Ophthalmology, Professor K. Gibinski University Clinical Center, Medical University of Silesia, Katowice, Poland
- Department of Ophthalmology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Ceglana 35, 40-514, Katowice, Poland
| | - Mariola Dorecka
- Department of Ophthalmology, Professor K. Gibinski University Clinical Center, Medical University of Silesia, Katowice, Poland
- Department of Ophthalmology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Ceglana 35, 40-514, Katowice, Poland
| | - Adrian Smedowski
- GlaucoTech Co, Katowice, Poland.
- Department of Ophthalmology, Professor K. Gibinski University Clinical Center, Medical University of Silesia, Katowice, Poland.
- Department of Ophthalmology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Ceglana 35, 40-514, Katowice, Poland.
| |
Collapse
|
4
|
Suzuki R, Woo JZ, Thumberger T, Hofmann G, Wittbrodt J, Tavhelidse-Suck T. Characterizing medaka visual features using a high-throughput optomotor response assay. PLoS One 2024; 19:e0302092. [PMID: 38941325 PMCID: PMC11213317 DOI: 10.1371/journal.pone.0302092] [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: 03/27/2024] [Accepted: 06/16/2024] [Indexed: 06/30/2024] Open
Abstract
Medaka fish (Oryzias latipes) is a powerful model to study genetics underlying the developmental and functional traits of the vertebrate visual system. We established a simple and high-throughput optomotor response (OMR) assay utilizing medaka larvae to study visual functions including visual acuity and contrast sensitivity. Our assay presents multiple adjustable stripes in motion to individual fish in a linear arena. For that the OMR assay employs a tablet display and the Fish Stripes software to adjust speed, width, color, and contrast of the stripes. Our results demonstrated that optomotor responses were robustly induced by black and white stripes presented from below in the linear-pool-arena. We detected robust strain specific differences in the OMR when comparing long established medaka inbred strains. We observed an interesting training effect upon the initial exposure of larvae to thick stripes, which allowed them to better respond to narrower stripes. The OMR setup and protocol presented here provide an efficient tool for quantitative phenotype mapping, addressing visual acuity, trainability of cortical neurons, color sensitivity, locomotor response, retinal regeneration and others. Our open-source setup presented here provides a crucial prerequisite for ultimately addressing the genetic basis of those processes.
Collapse
Affiliation(s)
- Risa Suzuki
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
- Heidelberg Biosciences International Graduate School (HBIGS), Heidelberg, Germany
| | - Jia Zheng Woo
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Thomas Thumberger
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Gero Hofmann
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | - Joachim Wittbrodt
- Centre for Organismal Studies (COS), Heidelberg University, Heidelberg, Germany
| | | |
Collapse
|
5
|
Arias-Montecino A, Sykes A, Álvarez-Hernán G, de Mera-Rodríguez JA, Calle-Guisado V, Martín-Partido G, Rodríguez-León J, Francisco-Morcillo J. Histological and scanning electron microscope observations on the developing retina of the cuttlefish (Sepia officinalis Linnaeus, 1758). Tissue Cell 2024; 88:102417. [PMID: 38820948 DOI: 10.1016/j.tice.2024.102417] [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: 01/29/2024] [Revised: 05/03/2024] [Accepted: 05/20/2024] [Indexed: 06/02/2024]
Abstract
In this work we present a detailed study of the major events during retinal histogenesis of the cuttlefish Sepia officinalis from early embryos to newly hatched animals and juveniles. For this purpose, we carried out morphometric and histological analyses using light and scanning electron microscopy. From St19, the first embryonic stage analysed, to St23/24 the embryonic retina is composed of a pseudostratified epithelium showing abundant mitotic figures in the more internal surface. At St24 the first photoreceptor nuclei appear in the presumptive inner segment layer, while an incipient layer of apical processes of the future rhabdomeric layer become visible at St25. From this stage onwards, both the rhabdomeric layer and the inner segment layer increase in size until postnatal ages. In contrast, the width of the supporting cell layer progressively decreases from St25/26 until postnatal ages. S. officinalis embryos hatched in a morphologically advanced state, showing a differentiated retina even in the last stages of the embryonic period. However, features of immaturity are still observable in the retinal tissue during the first postnatal weeks of life, such as the existence of mitotic figures in the apical region of the supporting cell layer and migrating nuclei of differentiating photoreceptors crossing the basal membrane to reach their final location in the inner segment layer. Therefore, postnatal retinal neurogenesis is present in juvenile specimens of S. officinalis.
Collapse
Affiliation(s)
- Alejandro Arias-Montecino
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, Badajoz 06006, Spain
| | - Antonio Sykes
- Center of Marine Sciences, Universidade do Algarve Campus de Gambelas, Faro 8005-139, Portugal
| | - Guadalupe Álvarez-Hernán
- Área de Anatomía y Embriología Humana, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Medicina, Universidad de Extremadura, Badajoz 06006, Spain.
| | - José Antonio de Mera-Rodríguez
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, Badajoz 06006, Spain
| | - Violeta Calle-Guisado
- Área de Anatomía y Embriología Humana, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Medicina, Universidad de Extremadura, Badajoz 06006, Spain
| | - Gervasio Martín-Partido
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, Badajoz 06006, Spain
| | - Joaquín Rodríguez-León
- Área de Anatomía y Embriología Humana, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Medicina, Universidad de Extremadura, Badajoz 06006, Spain
| | - Javier Francisco-Morcillo
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, Badajoz 06006, Spain
| |
Collapse
|
6
|
Zhang R, Sun J, Xie Y, Zhu W, Tao M, Chen Y, Xie W, Bade R, Jiang S, Liu X, Shao G, Pan W, Zhou C, Jia X. Mutant kri1l causes abnormal retinal development via cell cycle arrest and apoptosis induction. Cell Death Discov 2024; 10:251. [PMID: 38789412 PMCID: PMC11126728 DOI: 10.1038/s41420-024-02022-2] [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: 07/14/2023] [Revised: 05/04/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
Damage to the ribosome or an imbalance in protein biosynthesis can lead to some human diseases, such as diabetic retinopathy (DR) and other eye diseases. Here, we reported that the kri1l gene was responsible for retinal development. The kri1l gene encodes an essential component of the rRNA small subunit processome. The retinal structure was disrupted in kri1l mutants, which resulted in small eyes. The boundaries of each layer of cells in the retina were blurred, and each layer of cells was narrowed and decreased. The photoreceptor cells and Müller glia cells almost disappeared in kri1l mutants. The lack of photoreceptor cells caused a fear of light response. The development of the retina started without abnormalities, and the abnormalities began two days after fertilization. In the kri1l mutant, retinal cell differentiation was defective, resulting in the disappearance of cone cells and Müller cells. The proliferation of retinal cells was increased, while apoptosis was also enhanced in kri1l mutants. γ-H2AX upregulation indicated the accumulation of DNA damage, which resulted in cell cycle arrest and apoptosis. The kri1l mutation reduced the expression of some opsin genes and key retinal genes, which are also essential for retinal development.
Collapse
Affiliation(s)
- Rong Zhang
- Department of Basic Medicine and Forensic Medicine, Baotou Medical College, Inner Mongolia, Baotou, China
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, Baotou, China
- Fourth Hospital of Baotou, Inner Mongolia, Baotou, China
| | - Jiajun Sun
- Department of Basic Medicine and Forensic Medicine, Baotou Medical College, Inner Mongolia, Baotou, China
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, Baotou, China
| | - Yabin Xie
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, Baotou, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wei Zhu
- School of Pharmacy, Baotou Medical College, Inner Mongolia, Baotou, China
| | - Meitong Tao
- Department of Basic Medicine and Forensic Medicine, Baotou Medical College, Inner Mongolia, Baotou, China
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, Baotou, China
| | - Yu Chen
- Department of Basic Medicine and Forensic Medicine, Baotou Medical College, Inner Mongolia, Baotou, China
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, Baotou, China
| | - Wei Xie
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, Baotou, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Rengui Bade
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, Baotou, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shuyuan Jiang
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, Baotou, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- School of Pharmacy, Baotou Medical College, Inner Mongolia, Baotou, China
| | - Xiaolei Liu
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, Baotou, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- School of Pharmacy, Baotou Medical College, Inner Mongolia, Baotou, China
| | - Guo Shao
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, Baotou, China
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Center for Translational Medicine and Department of Laboratory Medicine, The Third People's Hospital of Longgang District, Shenzhen, China
| | - Weijun Pan
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Chengjiang Zhou
- Department of Basic Medicine and Forensic Medicine, Baotou Medical College, Inner Mongolia, Baotou, China.
| | - Xiaoe Jia
- Department of Basic Medicine and Forensic Medicine, Baotou Medical College, Inner Mongolia, Baotou, China.
- Inner Mongolia Key laboratory of Hypoxic Translational Medicine, Baotou Medical College, Inner Mongolia, Baotou, China.
- Beijing Key Laboratory of Hypoxic Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
7
|
Andreazzoli M, Longoni B, Angeloni D, Demontis GC. Retinoid Synthesis Regulation by Retinal Cells in Health and Disease. Cells 2024; 13:871. [PMID: 38786093 PMCID: PMC11120330 DOI: 10.3390/cells13100871] [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: 02/07/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
Vision starts in retinal photoreceptors when specialized proteins (opsins) sense photons via their covalently bonded vitamin A derivative 11cis retinaldehyde (11cis-RAL). The reaction of non-enzymatic aldehydes with amino groups lacks specificity, and the reaction products may trigger cell damage. However, the reduced synthesis of 11cis-RAL results in photoreceptor demise and suggests the need for careful control over 11cis-RAL handling by retinal cells. This perspective focuses on retinoid(s) synthesis, their control in the adult retina, and their role during retina development. It also explores the potential importance of 9cis vitamin A derivatives in regulating retinoid synthesis and their impact on photoreceptor development and survival. Additionally, recent advancements suggesting the pivotal nature of retinoid synthesis regulation for cone cell viability are discussed.
Collapse
Affiliation(s)
| | - Biancamaria Longoni
- Department of Translational Medicine and New Technologies in Medicine, University of Pisa, 56126 Pisa, Italy
| | - Debora Angeloni
- The Institute of Biorobotics, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | | |
Collapse
|
8
|
Gong Y, Ge L, Li Q, Gong J, Chen M, Gao H, Kang J, Yu T, Li J, Xu H. Ethanol Causes Cell Death and Neuronal Differentiation Defect During Initial Neurogenesis of the Neural Retina by Disrupting Calcium Signaling in Human Retinal Organoids. Stem Cell Rev Rep 2023; 19:2790-2806. [PMID: 37603136 DOI: 10.1007/s12015-023-10604-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2023] [Indexed: 08/22/2023]
Abstract
Fetal Alcohol Syndrome (FAS) affects a significant proportion, exceeding 90%, of afflicted children, leading to severe ocular aberrations such as microphthalmia and optic nerve hypoplasia. During the early stages of pregnancy, the commencement of neural retina neurogenesis represents a critical period for human eye development, concurrently exposing the developing retinal structures to the highest risk of prenatal ethanol exposure due to a lack of awareness. Despite the paramount importance of this period, the precise influence and underlying mechanisms of short-term ethanol exposure on the developmental process of the human neural retina have remained largely elusive. In this study, we utilize the human embryonic stem cells derived retinal organoids (hROs) to recapitulate the initial retinal neurogenesis and find that 1% (v/v) ethanol slows the growth of hROs by inducing robust cell death and retinal ganglion cell differentiation defect. Bulk RNA-seq analysis and two-photon microscope live calcium imaging reveal altered calcium signaling dynamics derived from ethanol-induced down-regulation of RYR1 and CACNA1S. Moreover, the calcium-binding protein RET, one of the downstream effector genes of the calcium signaling pathway, synergistically integrates ethanol and calcium signals to abort neuron differentiation and cause cell death. To sum up, our study illustrates the effect and molecular mechanism of ethanol on the initial neurogenesis of the human embryonic neural retina, providing a novel interpretation of the ocular phenotype of FAS and potentially informing preventative measures for susceptible populations.
Collapse
Affiliation(s)
- Yu Gong
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
- Department of Ophthalmology, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Lingling Ge
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
| | - Qiyou Li
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
| | - Jing Gong
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Min Chen
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
| | - Hui Gao
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
| | - Jiahui Kang
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China
| | - Ting Yu
- Department of Clinical Laboratory, The 89th Hospital of The People's Liberation Army, Weifang, People's Republic of China
| | - Jiawen Li
- Department of Ophthalmology, University-Town Hospital of Chongqing Medical University, Chongqing, China.
| | - Haiwei Xu
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, People's Republic of China.
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, People's Republic of China.
| |
Collapse
|
9
|
Lathe R, St Clair D. Programmed ageing: decline of stem cell renewal, immunosenescence, and Alzheimer's disease. Biol Rev Camb Philos Soc 2023; 98:1424-1458. [PMID: 37068798 DOI: 10.1111/brv.12959] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/19/2023]
Abstract
The characteristic maximum lifespan varies enormously across animal species from a few hours to hundreds of years. This argues that maximum lifespan, and the ageing process that itself dictates lifespan, are to a large extent genetically determined. Although controversial, this is supported by firm evidence that semelparous species display evolutionarily programmed ageing in response to reproductive and environmental cues. Parabiosis experiments reveal that ageing is orchestrated systemically through the circulation, accompanied by programmed changes in hormone levels across a lifetime. This implies that, like the circadian and circannual clocks, there is a master 'clock of age' (circavital clock) located in the limbic brain of mammals that modulates systemic changes in growth factor and hormone secretion over the lifespan, as well as systemic alterations in gene expression as revealed by genomic methylation analysis. Studies on accelerated ageing in mice, as well as human longevity genes, converge on evolutionarily conserved fibroblast growth factors (FGFs) and their receptors, including KLOTHO, as well as insulin-like growth factors (IGFs) and steroid hormones, as key players mediating the systemic effects of ageing. Age-related changes in these and multiple other factors are inferred to cause a progressive decline in tissue maintenance through failure of stem cell replenishment. This most severely affects the immune system, which requires constant renewal from bone marrow stem cells. Age-related immune decline increases risk of infection whereas lifespan can be extended in germfree animals. This and other evidence suggests that infection is the major cause of death in higher organisms. Immune decline is also associated with age-related diseases. Taking the example of Alzheimer's disease (AD), we assess the evidence that AD is caused by immunosenescence and infection. The signature protein of AD brain, Aβ, is now known to be an antimicrobial peptide, and Aβ deposits in AD brain may be a response to infection rather than a cause of disease. Because some cognitively normal elderly individuals show extensive neuropathology, we argue that the location of the pathology is crucial - specifically, lesions to limbic brain are likely to accentuate immunosenescence, and could thus underlie a vicious cycle of accelerated immune decline and microbial proliferation that culminates in AD. This general model may extend to other age-related diseases, and we propose a general paradigm of organismal senescence in which declining stem cell proliferation leads to programmed immunosenescence and mortality.
Collapse
Affiliation(s)
- Richard Lathe
- Division of Infection Medicine, Chancellor's Building, University of Edinburgh Medical School, Little France, Edinburgh, EH16 4SB, UK
| | - David St Clair
- Institute of Medical Sciences, School of Medicine, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| |
Collapse
|
10
|
Sokolova N, Zilova L, Wittbrodt J. Unravelling the link between embryogenesis and adult stem cell potential in the ciliary marginal zone: A comparative study between mammals and teleost fish. Cells Dev 2023; 174:203848. [PMID: 37172718 DOI: 10.1016/j.cdev.2023.203848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
The discovery and study of adult stem cells have revolutionized regenerative medicine by offering new opportunities for treating various medical conditions. Anamniote stem cells, which retain their full proliferative capacity and full differentiation range throughout their lifetime, harbour a greater potential compared to mammalian adult stem cells, which only exhibit limited stem cell potential. Therefore, understanding the mechanisms underlying these differences is of significant interest. In this review, we examine the similarities and differences of adult retinal stem cells in anamniotes and mammals, from their embryonic stages in the optic vesicle to their residence in the postembryonic retinal stem cell niche, the ciliary marginal zone located in the retinal periphery. In anamniotes, developing precursors of retinal stem cells are exposed to various environmental cues during their migration in the complex morphogenetic remodelling of the optic vesicle to the optic cup. In contrast, their mammalian counterparts in the retinal periphery are primarily instructed by neighbouring tissues once they are in place. We explore the distinct modes of optic cup morphogenesis in mammals and teleost fish and highlight molecular mechanisms governing morphogenesis and stem cells instruction. The review concludes with the molecular mechanisms of ciliary marginal zone formation and offers a perspective on the impact of comparative single cell transcriptomic studies to reveal the evolutionary similarities and differences.
Collapse
Affiliation(s)
- Natalia Sokolova
- Centre for Organismal Studies Heidelberg, Germany; Heidelberg Biosciences International Graduate School, Germany
| | - Lucie Zilova
- Centre for Organismal Studies Heidelberg, Germany.
| | | |
Collapse
|
11
|
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: 13] [Impact Index Per Article: 6.5] [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
|
12
|
Qiu C, Martin BK, Welsh IC, Daza RM, Le TM, Huang X, Nichols EK, Taylor ML, Fulton O, O’Day DR, Gomes AR, Ilcisin S, Srivatsan S, Deng X, Disteche CM, Noble WS, Hamazaki N, Moens CB, Kimelman D, Cao J, Schier AF, Spielmann M, Murray SA, Trapnell C, Shendure J. A single-cell transcriptional timelapse of mouse embryonic development, from gastrula to pup. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.05.535726. [PMID: 37066300 PMCID: PMC10104014 DOI: 10.1101/2023.04.05.535726] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
The house mouse, Mus musculus, is an exceptional model system, combining genetic tractability with close homology to human biology. Gestation in mouse development lasts just under three weeks, a period during which its genome orchestrates the astonishing transformation of a single cell zygote into a free-living pup composed of >500 million cells. Towards a global framework for exploring mammalian development, we applied single cell combinatorial indexing (sci-*) to profile the transcriptional states of 12.4 million nuclei from 83 precisely staged embryos spanning late gastrulation (embryonic day 8 or E8) to birth (postnatal day 0 or P0), with 2-hr temporal resolution during somitogenesis, 6-hr resolution through to birth, and 20-min resolution during the immediate postpartum period. From these data (E8 to P0), we annotate dozens of trajectories and hundreds of cell types and perform deeper analyses of the unfolding of the posterior embryo during somitogenesis as well as the ontogenesis of the kidney, mesenchyme, retina, and early neurons. Finally, we leverage the depth and temporal resolution of these whole embryo snapshots, together with other published data, to construct and curate a rooted tree of cell type relationships that spans mouse development from zygote to pup. Throughout this tree, we systematically nominate sets of transcription factors (TFs) and other genes as candidate drivers of the in vivo differentiation of hundreds of mammalian cell types. Remarkably, the most dramatic shifts in transcriptional state are observed in a restricted set of cell types in the hours immediately following birth, and presumably underlie the massive changes in physiology that must accompany the successful transition of a placental mammal to extrauterine life.
Collapse
Affiliation(s)
- Chengxiang Qiu
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Beth K. Martin
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | | | - Riza M. Daza
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Truc-Mai Le
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Xingfan Huang
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA, USA
| | - Eva K. Nichols
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Megan L. Taylor
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Olivia Fulton
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Diana R. O’Day
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | | | - Saskia Ilcisin
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
| | - Sanjay Srivatsan
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Xinxian Deng
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Christine M. Disteche
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - William Stafford Noble
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, WA, USA
| | - Nobuhiko Hamazaki
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
| | - Cecilia B. Moens
- Division of Basic Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - David Kimelman
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Junyue Cao
- Laboratory of Single-cell genomics and Population dynamics, The Rockefeller University, New York, NY, USA
| | - Alexander F. Schier
- Biozentrum, University of Basel, Basel, Switzerland
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| | - Malte Spielmann
- Max Planck Institute for Molecular Genetics, Berlin, Germany
- Institute of Human Genetics, University Hospitals Schleswig-Holstein, University of Lübeck and Kiel University, Lübeck, Kiel, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Hamburg, Lübeck, Kiel, Lübeck, Germany
| | | | - Cole Trapnell
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
| |
Collapse
|
13
|
Álvarez-Hernán G, de Mera-Rodríguez JA, Calle-Guisado V, Martín-Partido G, Rodríguez-León J, Francisco-Morcillo J. Retinal Development in a Precocial Bird Species, the Quail (Coturnix coturnix, Linnaeus 1758). Cells 2023; 12:cells12070989. [PMID: 37048062 PMCID: PMC10093483 DOI: 10.3390/cells12070989] [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: 01/25/2023] [Revised: 03/09/2023] [Accepted: 03/15/2023] [Indexed: 03/28/2023] Open
Abstract
The quail (Coturnix coturnix, Linnaeus 1758), a notable model used in developmental biology, is a precocial bird species in which the processes of retinal cell differentiation and retinal histogenesis have been poorly studied. The purpose of the present research is to examine the retinogenesis in this bird species immunohistochemically and compare the results with those from previous studies in precocial and altricial birds. We found that the first PCNA-negative nuclei are detected at Stage (St) 21 in the vitreal region of the neuroblastic layer, coinciding topographically with the first αTubAc-/Tuj1-/Isl1-immunoreactive differentiating ganglion cells. At St28, the first Prox1-immunoreactive nuclei can be distinguished in the vitreal side of the neuroblastic layer (NbL), but also the first visinin-immunoreactive photoreceptors in the scleral surface. The inner plexiform layer (IPL) emerges at St32, and the outer plexiform layer (OPL) becomes visible at St35—the stage in which the first GS-immunoreactive Müller cells are distinguishable. Newly hatched animals show a well-developed stratified retina in which the PCNA-and pHisH3-immunoreactivies are absent. Therefore, retinal cell differentiation in the quail progresses in the stereotyped order conserved among vertebrates, in which ganglion cells initially appear and are followed by amacrine cells, horizontal cells, and photoreceptors. Müller glia are one of the last cell types to be born. Plexiform layers emerge following a vitreal-to-scleral gradient. Finally, our results suggest that there are no significant differences in the timing of different events involved in retinal maturation between the quail and the chicken, but the same events are delayed in an altricial bird species.
Collapse
Affiliation(s)
- Guadalupe Álvarez-Hernán
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, 06006 Badajoz, Spain
| | - José Antonio de Mera-Rodríguez
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, 06006 Badajoz, Spain
| | - Violeta Calle-Guisado
- Área de Anatomía y Embriología Humana, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Medicina, Universidad de Extremadura, 06006 Badajoz, Spain
| | - Gervasio Martín-Partido
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, 06006 Badajoz, Spain
| | - Joaquín Rodríguez-León
- Área de Anatomía y Embriología Humana, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Medicina, Universidad de Extremadura, 06006 Badajoz, Spain
| | - Javier Francisco-Morcillo
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, 06006 Badajoz, Spain
- Correspondence:
| |
Collapse
|
14
|
Arthur P, Muok L, Nathani A, Zeng EZ, Sun L, Li Y, Singh M. Bioengineering Human Pluripotent Stem Cell-Derived Retinal Organoids and Optic Vesicle-Containing Brain Organoids for Ocular Diseases. Cells 2022; 11:3429. [PMID: 36359825 PMCID: PMC9653705 DOI: 10.3390/cells11213429] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/13/2022] [Accepted: 10/23/2022] [Indexed: 08/24/2023] Open
Abstract
Retinal organoids are three-dimensional (3D) structures derived from human pluripotent stem cells (hPSCs) that mimic the retina's spatial and temporal differentiation, making them useful as in vitro retinal development models. Retinal organoids can be assembled with brain organoids, the 3D self-assembled aggregates derived from hPSCs containing different cell types and cytoarchitectures that resemble the human embryonic brain. Recent studies have shown the development of optic cups in brain organoids. The cellular components of a developing optic vesicle-containing organoids include primitive corneal epithelial and lens-like cells, retinal pigment epithelia, retinal progenitor cells, axon-like projections, and electrically active neuronal networks. The importance of retinal organoids in ocular diseases such as age-related macular degeneration, Stargardt disease, retinitis pigmentosa, and diabetic retinopathy are described in this review. This review highlights current developments in retinal organoid techniques, and their applications in ocular conditions such as disease modeling, gene therapy, drug screening and development. In addition, recent advancements in utilizing extracellular vesicles secreted by retinal organoids for ocular disease treatments are summarized.
Collapse
Affiliation(s)
- Peggy Arthur
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Laureana Muok
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32306, USA
| | - Aakash Nathani
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Eric Z. Zeng
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32306, USA
| | - Li Sun
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32306, USA
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32306, USA
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| |
Collapse
|
15
|
Hiramatsu N, Yamamoto N, Ohkuma M, Nagai N, Miyachi EI, Yamatsuta K, Imaizumi K. Iris-derived induced pluripotent stem cells that express GFP in all somatic cells of mice and differentiate into functional retinal neurons. Med Mol Morphol 2022; 55:292-303. [PMID: 35932315 DOI: 10.1007/s00795-022-00330-z] [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: 05/02/2022] [Accepted: 06/29/2022] [Indexed: 11/25/2022]
Abstract
When regenerated tissue is generated from induced pluripotent stem cells (iPSCs), it is necessary to track and identify the transplanted cells. Fluorescently-labeled iPSCs synthesize a fluorescent substance that is easily tracked. However, the expressed protein should not affect the original genome sequence or pluripotency. To solve this problem, we created a cell tool for basic research on iPSCs. Iris tissue-derived cells from GFP fluorescence-expressing mice (GFP-DBA/2 mice) were reprogrammed to generate GFP mouse iris-derived iPSCs (M-iris GFP iPSCs). M-iris GFP iPSCs expressed cell markers characteristic of iPSCs and showed pluripotency in differentiating into the three germ layers. In addition, when expressing GFP, the cells differentiated into functional recoverin- and calbindin-positive cells. Thus, this cell line will facilitate future studies on iPSCs.
Collapse
Affiliation(s)
- Noriko Hiramatsu
- Support Office for Bioresource Research, Research Promotion Headquarters, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
- Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Naoki Yamamoto
- Support Office for Bioresource Research, Research Promotion Headquarters, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan.
- International Center for Cell and Gene Therapy, Research Promotion and Support Headquarters, Fujita Health University, Toyoake, Aichi, 470-1192, Japan.
- Department of Ophthalmology, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan.
- Division of Vision Research for Environmental Health, Project Research Center, Medical Research Institute, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan.
| | - Mahito Ohkuma
- Department of Physiology, School of Medicine, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Noriaki Nagai
- Faculty of Pharmacy, Kindai University, Higashiosaka, Osaka, 577-8502, Japan
| | - Ei-Ichi Miyachi
- Department of Physiology, School of Medicine, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
- Department of Food Science and Nutrition, Nagoya Women's University, Toyoake, Aichi, 467-8610, Japan
| | - Kumiko Yamatsuta
- Department of Respiratory Medicine, Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan
| | - Kazuyoshi Imaizumi
- Department of Respiratory Medicine, Fujita Health University School of Medicine, Toyoake, Aichi, 470-1192, Japan
| |
Collapse
|
16
|
Huang YS, Lin CY. Stimulatory Effects of Androgens on Eel Primary Ovarian Development - from Phenotypes to Genotypes. Vet Med Sci 2022. [DOI: 10.5772/intechopen.99582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Androgens stimulate primary ovarian development in Vertebrate. Japanese eels underwent operation to sample the pre- and post-treated ovarian tissues from the same individual. Ovarian phenotypic or genotypic data were mined in a pair. A correlation between the initial ovarian status (determined by kernel density estimation (KDE), presented as a probability density of oocyte size) and the consequence of androgen (17MT) treatment (change in ovary) has been showed. The initial ovarian status appeared to be important to influence ovarian androgenic sensitivity. The initial ovary was important to the outcomes of androgen treatments, and ePAV (expression presence-absence variation) is existing in Japanese eel by analyze DEGs; core, unique, or accessory genes were identified, the sensitivities of initial ovaries were correlated with their gene expression profiles. We speculated the importance of genetic differential expression on the variations of phenotypes by 17MT, and transcriptomic approach seems to allow extracting multiple layers of genomic data.
Collapse
|
17
|
Li C, Shi X, Yang J, Li K, Dai L, Zhang Y, Zhou M, Su J. Genome-wide characterization of RNA editing highlights roles of high editing events of glutamatergic synapse during mouse retinal development. Comput Struct Biotechnol J 2022; 20:2648-2656. [PMID: 35685368 PMCID: PMC9162912 DOI: 10.1016/j.csbj.2022.05.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/14/2022] [Accepted: 05/14/2022] [Indexed: 11/30/2022] Open
Abstract
Adenosine-to-inosine (A-to-I) RNA editing leads to functional change of neurotransmitter receptor which is essential for neurotransmission and normal neuronal development. As a highly accessible part of central nervous system, retina has been extensively studied, however, it remains largely unknown how RNA editing regulates its development. Here, a genome-wide screening of high-confidence RNA editing events were performed to decipher the dynamic transcriptome regulation by RNA editing during mouse retinal development. 2000 high-confidence editing sites across eight developmental stages of retina were called. Three unique patterns (RNA-editinghigh pattern, RNA-editingmedium pattern and RNA-editinglow pattern) were identified by clustering these editing sites based on their editing level during retinal development. Editing events from RNA-editinghigh pattern were significantly associated with glutamate receptors and regulated synaptic transmission. Interestingly, most non-synonymous high-editing sites were mapped to ion channel genes of glutamatergic synapse which were associated with neurotransmission by controlling ion channel permeability and affecting exocytosis. Meanwhile, these non-synonymous editing sites were evolutionarily conserved and exhibited a consistently increasing editing levels between mouse and human retinal development. Single-cell RNA-seq data analysis revealed that RNA editing events prefer to occur in two main cell types including bipolar and amacrine cells. Genes with non-synonymous high-editing sites were enriched in both bipolar cells and retina ganglion cells, which may mediate retina ganglion cell differentiation by altering channel ion permeability. Together, our results provide novel insights into mechanism of post-transcriptional regulation during retinal development and help to develop novel RNA editing-guided therapeutic strategies for retinal disorders.
Collapse
Affiliation(s)
- Chenghao Li
- School of Biomedical Engineering, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325001, China
| | - Xinrui Shi
- School of Biomedical Engineering, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jiaying Yang
- School of Biomedical Engineering, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Ke Li
- School of Biomedical Engineering, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Lijun Dai
- School of Biomedical Engineering, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yan Zhang
- School of Biomedical Engineering, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Meng Zhou
- School of Biomedical Engineering, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jianzhong Su
- School of Biomedical Engineering, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang 325001, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325011, China
| |
Collapse
|
18
|
Wang X, Sarver AL, Han Q, Seiler CL, Xie C, Lu H, Forster CL, Tretyakova NY, Hallstrom TC. UHRF2 regulates cell cycle, epigenetics and gene expression to control the timing of retinal progenitor and ganglion cell differentiation. Development 2022; 149:274710. [PMID: 35285483 PMCID: PMC8984156 DOI: 10.1242/dev.195644] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 01/28/2022] [Indexed: 11/20/2022]
Abstract
Ubiquitin-like, containing PHD and RING finger domains 2 (UHRF2) regulates cell cycle and binds 5-hydroxymethylcytosine (5hmC) to promote completion of DNA demethylation. Uhrf2-/- mice are without gross phenotypic defects; however, the cell cycle and epigenetic regulatory functions of Uhrf2 during retinal tissue development are unclear. Retinal progenitor cells (RPCs) produce all retinal neurons and Müller glia in a predictable sequence controlled by the complex interplay between extrinsic signaling, cell cycle, epigenetic changes and cell-specific transcription factor activation. In this study, we find that UHRF2 accumulates in RPCs, and its conditional deletion from mouse RPCs reduced 5hmC, altered gene expressions and disrupted retinal cell proliferation and differentiation. Retinal ganglion cells were overproduced in Uhrf2-deficient retinae at the expense of VSX2+ RPCs. Most other cell types were transiently delayed in differentiation. Expression of each member of the Tet3/Uhrf2/Tdg active demethylation pathway was reduced in Uhrf2-deficient retinae, consistent with locally reduced 5hmC in their gene bodies. This study highlights a novel role of UHRF2 in controlling the transition from RPCs to differentiated cell by regulating cell cycle, epigenetic and gene expression decisions.
Collapse
Affiliation(s)
- Xiaohong Wang
- Department of Pediatrics, Division of Blood and Marrow Transplantation, 420 Delaware Street S.E., University of Minnesota, Minneapolis, MN 55455, USA
| | - Aaron L Sarver
- Institute for Health Informatics, Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Qiyuan Han
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Christopher L Seiler
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Chencheng Xie
- Department of Pediatrics, Division of Blood and Marrow Transplantation, 420 Delaware Street S.E., University of Minnesota, Minneapolis, MN 55455, USA
| | - Huarui Lu
- Department of Pediatrics, Division of Blood and Marrow Transplantation, 420 Delaware Street S.E., University of Minnesota, Minneapolis, MN 55455, USA
| | - Colleen L Forster
- BioNet, Academic Health Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Natalia Y Tretyakova
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Timothy C Hallstrom
- Department of Pediatrics, Division of Blood and Marrow Transplantation, 420 Delaware Street S.E., University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
19
|
Bradshaw SN, Allison WT. Hagfish to Illuminate the Developmental and Evolutionary Origins of the Vertebrate Retina. Front Cell Dev Biol 2022; 10:822358. [PMID: 35155434 PMCID: PMC8826474 DOI: 10.3389/fcell.2022.822358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/07/2022] [Indexed: 11/13/2022] Open
Abstract
The vertebrate eye is a vital sensory organ that has long fascinated scientists, but the details of how this organ evolved are still unclear. The vertebrate eye is distinct from the simple photoreceptive organs of other non-vertebrate chordates and there are no clear transitional forms of the eye in the fossil record. To investigate the evolution of the eye we can examine the eyes of the most ancient extant vertebrates, the hagfish and lamprey. These jawless vertebrates are in an ideal phylogenetic position to study the origin of the vertebrate eye but data on eye/retina development in these organisms is limited. New genomic and gene expression data from hagfish and lamprey suggest they have many of the same genes for eye development and retinal neurogenesis as jawed vertebrates, but functional work to determine if these genes operate in retinogenesis similarly to other vertebrates is missing. In addition, hagfish express a marker of proliferative retinal cells (Pax6) near the margin of the retina, and adult retinal growth is apparent in some species. This finding of eye growth late into hagfish ontogeny is unexpected given the degenerate eye phenotype. Further studies dissecting retinal neurogenesis in jawless vertebrates would allow for comparison of the mechanisms of retinal development between cyclostome and gnathostome eyes and provide insight into the evolutionary origins of the vertebrate eye.
Collapse
Affiliation(s)
| | - W. Ted Allison
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
20
|
Too LK, Simunovic MP. Retinal Stem/Progenitor Cells Derived From Adult Müller Glia for the Treatment of Retinal Degeneration. Front Cell Dev Biol 2021; 9:749131. [PMID: 34660607 PMCID: PMC8511496 DOI: 10.3389/fcell.2021.749131] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/06/2021] [Indexed: 01/09/2023] Open
Abstract
Over the past two decades, progress in our understanding of glial function has been revolutionary. Within the retina, a subset of glial cells termed the “Müller glia (MG),” have been demonstrated to play key roles in retinal homeostasis, structure and metabolism. Additionally, MG have also been shown to possess the regenerative capacity that varies across species. In teleost fish, MG respond to injury by reprogramming into stem-like cells capable of regenerating lost tissue. The expression of stem/progenitor cell markers has been demonstrated broadly in mammalian MG, including human MG, but their in vivo regenerative capacity appears evolutionarily limited. Advances in stem cell therapy have progressively elucidated critical mechanisms underlying innate MG reprogramming in teleost fish, which have shown promising results when applied to rodents. Furthermore, when cultured ex vivo, MG from mammals can differentiate into several retina cell types. In this review, we will explore the reparative and regenerative potential of MG in cellular therapy approaches, and outline our current understanding of embryonic retinal development, the stem-cell potential of MG in adult vertebrate retina (including human), and microenvironmental cues that guide MG reprogramming.
Collapse
Affiliation(s)
- Lay Khoon Too
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Matthew P Simunovic
- Save Sight Institute, The University of Sydney, Sydney, NSW, Australia.,Sydney Eye Hospital, Sydney, NSW, Australia
| |
Collapse
|
21
|
Villarejo-Zori B, Jiménez-Loygorri JI, Zapata-Muñoz J, Bell K, Boya P. New insights into the role of autophagy in retinal and eye diseases. Mol Aspects Med 2021; 82:101038. [PMID: 34620506 DOI: 10.1016/j.mam.2021.101038] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/12/2021] [Accepted: 09/27/2021] [Indexed: 02/06/2023]
Abstract
Autophagy is a fundamental homeostatic pathway that mediates the degradation and recycling of intracellular components. It serves as a key quality control mechanism, especially in non-dividing cells such as neurons. Proteins, lipids, and even whole organelles are engulfed in autophagosomes and delivered to the lysosome for elimination. The retina is a light-sensitive tissue located in the back of the eye that detects and processes visual images. Vision is a highly demanding process, making the eye one of the most metabolically active tissues in the body and photoreceptors display glycolytic metabolism, even in the presence of oxygen. The retina and eye are also exposed to other stressors that can impair their function, including genetic mutations and age-associated changes. Autophagy, among other pathways, is therefore a key process for the preservation of retinal homeostasis. Here, we review the roles of both canonical and non-canonical autophagy in normal retinal function. We discuss the most recent studies investigating the participation of autophagy in eye diseases such as age-related macular degeneration, glaucoma, and diabetic retinopathy and its role protecting photoreceptors in several forms of retinal degeneration. Finally, we consider the therapeutic potential of strategies that target autophagy pathways to treat prevalent retinal and eye diseases.
Collapse
Affiliation(s)
- Beatriz Villarejo-Zori
- Department of Cellular and Molecular Biology, Margarita Salas Center for Biological Research, CSIC, Ramiro de Maetzu, 9, 28040, Madrid, Spain
| | - Juan Ignacio Jiménez-Loygorri
- Department of Cellular and Molecular Biology, Margarita Salas Center for Biological Research, CSIC, Ramiro de Maetzu, 9, 28040, Madrid, Spain
| | - Juan Zapata-Muñoz
- Department of Cellular and Molecular Biology, Margarita Salas Center for Biological Research, CSIC, Ramiro de Maetzu, 9, 28040, Madrid, Spain
| | - Katharina Bell
- Singapore Eye Research Institute, Singapore National Eye Centre, Republic of Singapore
| | - Patricia Boya
- Department of Cellular and Molecular Biology, Margarita Salas Center for Biological Research, CSIC, Ramiro de Maetzu, 9, 28040, Madrid, Spain.
| |
Collapse
|
22
|
Guy B, Zhang JS, Duncan LH, Johnston RJ. Human neural organoids: Models for developmental neurobiology and disease. Dev Biol 2021; 478:102-121. [PMID: 34181916 PMCID: PMC8364509 DOI: 10.1016/j.ydbio.2021.06.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/08/2021] [Accepted: 06/24/2021] [Indexed: 12/25/2022]
Abstract
Human organoids stand at the forefront of basic and translational research, providing experimentally tractable systems to study human development and disease. These stem cell-derived, in vitro cultures can generate a multitude of tissue and organ types, including distinct brain regions and sensory systems. Neural organoid systems have provided fundamental insights into molecular mechanisms governing cell fate specification and neural circuit assembly and serve as promising tools for drug discovery and understanding disease pathogenesis. In this review, we discuss several human neural organoid systems, how they are generated, advances in 3D imaging and bioengineering, and the impact of organoid studies on our understanding of the human nervous system.
Collapse
Affiliation(s)
- Brian Guy
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD, 21218, USA
| | - Jingliang Simon Zhang
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD, 21218, USA
| | - Leighton H Duncan
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Robert J Johnston
- Department of Biology, Johns Hopkins University, 3400 N. Charles Street, Baltimore, MD, 21218, USA.
| |
Collapse
|
23
|
Pérez-Dones D, Ledesma-Terrón M, Míguez DG. Quantitative Approaches to Study Retinal Neurogenesis. Biomedicines 2021; 9:1222. [PMID: 34572408 PMCID: PMC8471905 DOI: 10.3390/biomedicines9091222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/07/2021] [Accepted: 09/11/2021] [Indexed: 11/16/2022] Open
Abstract
The study of the development of the vertebrate retina can be addressed from several perspectives: from a purely qualitative to a more quantitative approach that takes into account its spatio-temporal features, its three-dimensional structure and also the regulation and properties at the systems level. Here, we review the ongoing transition toward a full four-dimensional characterization of the developing vertebrate retina, focusing on the challenges at the experimental, image acquisition, image processing and quantification. Using the developing zebrafish retina, we illustrate how quantitative data extracted from these type of highly dense, three-dimensional tissues depend strongly on the image quality, image processing and algorithms used to segment and quantify. Therefore, we propose that the scientific community that focuses on developmental systems could strongly benefit from a more detailed disclosure of the tools and pipelines used to process and analyze images from biological samples.
Collapse
Affiliation(s)
- Diego Pérez-Dones
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Física de la Materia Condensada (IFIMAC), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Mario Ledesma-Terrón
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Física de la Materia Condensada (IFIMAC), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - David G Míguez
- Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, 28049 Madrid, Spain
- Física de la Materia Condensada (IFIMAC), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
24
|
Döpper H, Menges J, Bozet M, Brenzel A, Lohmann D, Steenpass L, Kanber D. Differentiation Protocol for 3D Retinal Organoids, Immunostaining and Signal Quantitation. ACTA ACUST UNITED AC 2021; 55:e120. [PMID: 32956559 DOI: 10.1002/cpsc.120] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Structures resembling whole organs, called organoids, are generated using pluripotent stem cells and 3D culturing methods. This relies on the ability of cells to self-reorganize after dissociation. In combination with certain supplemented factors, differentiation can be directed toward the formation of several organ-like structures. Here, a protocol for the generation of retinal organoids containing all seven retinal cell types is described. This protocol does not depend on Matrigel, and by keeping the organoids single and independent at all times, fusion is prevented and monitoring of differentiation is improved. Comprehensive phenotypic characterization of the in vitro-generated retinal organoids is achieved by the protocol for immunostaining outlined here. By comparing different stages of retinal organoids, the decrease and increase of certain cell populations can be determined. In order to be able to detect even small differences, it is necessary to quantify the immunofluorescent signals, for which we have provided a detailed protocol describing signal quantitation using the image-processing program Fiji. © 2020 The Authors. Basic Protocol 1: Differentiation protocol for 3D retinal organoids Basic Protocol 2: Immunostaining protocol for cryosections of retinal organoids Support Protocol: Embedding and sectioning protocol for 3D retinal organoids Basic Protocol 3: Quantitation protocol using Fiji.
Collapse
Affiliation(s)
- Hannah Döpper
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Julia Menges
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Morgane Bozet
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Alexandra Brenzel
- Institute for Experimental Immunology and Imaging, Imaging Center Essen (LMU), University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dietmar Lohmann
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Laura Steenpass
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Present address: Department of Human and Animal Cell Lines, Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures GmbH, Braunschweig, Germany
| | - Deniz Kanber
- Institute of Human Genetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
25
|
Wagstaff EL, Heredero Berzal A, Boon CJF, Quinn PMJ, ten Asbroek ALMA, Bergen AA. The Role of Small Molecules and Their Effect on the Molecular Mechanisms of Early Retinal Organoid Development. Int J Mol Sci 2021; 22:7081. [PMID: 34209272 PMCID: PMC8268497 DOI: 10.3390/ijms22137081] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/23/2021] [Accepted: 06/26/2021] [Indexed: 12/12/2022] Open
Abstract
Early in vivo embryonic retinal development is a well-documented and evolutionary conserved process. The specification towards eye development is temporally controlled by consecutive activation or inhibition of multiple key signaling pathways, such as the Wnt and hedgehog signaling pathways. Recently, with the use of retinal organoids, researchers aim to manipulate these pathways to achieve better human representative models for retinal development and disease. To achieve this, a plethora of different small molecules and signaling factors have been used at various time points and concentrations in retinal organoid differentiations, with varying success. Additions differ from protocol to protocol, but their usefulness or efficiency has not yet been systematically reviewed. Interestingly, many of these small molecules affect the same and/or multiple pathways, leading to reduced reproducibility and high variability between studies. In this review, we make an inventory of the key signaling pathways involved in early retinogenesis and their effect on the development of the early retina in vitro. Further, we provide a comprehensive overview of the small molecules and signaling factors that are added to retinal organoid differentiation protocols, documenting the molecular and functional effects of these additions. Lastly, we comparatively evaluate several of these factors using our established retinal organoid methodology.
Collapse
Affiliation(s)
- Ellie L. Wagstaff
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands;
| | - Andrea Heredero Berzal
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands; (A.H.B.); (C.J.F.B.)
| | - Camiel J. F. Boon
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands; (A.H.B.); (C.J.F.B.)
- Department of Ophthalmology, Leiden University Medical Center (LUMC), 2333 ZA Leiden, The Netherlands
| | - Peter M. J. Quinn
- Jonas Children’s Vision Care and Bernard & Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology & Cell Biology, Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center—New York-Presbyterian Hospital, New York, NY 10032, USA;
| | | | - Arthur A. Bergen
- Department of Human Genetics, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands;
- Department of Ophthalmology, Amsterdam UMC, University of Amsterdam (UvA), 1105 AZ Amsterdam, The Netherlands; (A.H.B.); (C.J.F.B.)
- Netherlands Institute for Neuroscience (NIN-KNAW), 1105 BA Amsterdam, The Netherlands
| |
Collapse
|
26
|
Huang YS, Cheng WC, Lin CY. Androgenic Sensitivities and Ovarian Gene Expression Profiles Prior to Treatment in Japanese Eel (Anguilla japonica). MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2021; 23:430-444. [PMID: 34191211 DOI: 10.1007/s10126-021-10035-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 04/28/2021] [Indexed: 06/13/2023]
Abstract
Androgens stimulate ovarian development in eels. Our previous report indicated a correlation between the initial (debut) ovarian status (determined by kernel density estimation (KDE), presented as a probability density of oocyte size) and the consequence of 17MT treatment (change in ovary). The initial ovarian status appeared to be an important factor influencing ovarian androgenic sensitivity. We postulated that the sensitivities of initial ovaries are correlated with their gene expression profiles. Japanese eels underwent operation to sample the initial ovarian tissues, and the samples were stored in liquid nitrogen. Using high-throughput next-generation sequencing (NGS) technology, ovarian transcriptomic data were mined and analyzed based on functional gene classification with cutoff-based differentially expressed genes (DEGs); the ovarian status was transformed into gene expression profiles globally or was represented by a set of gene list. Our results also implied that the initial ovary might be an important factor influencing the outcomes of 17MT treatments, and the genes related with neuronal activities or neurogenesis seemed to play an essential role in the positive effect.
Collapse
Affiliation(s)
- Yung-Sen Huang
- Department of Life Science, National University of Kaohsiung, No. 700 Kaohsiung University Road, Nan Tzu Dist, 811, Kaohsiung, Taiwan.
| | - Wen-Chih Cheng
- Institute of Information Science, Academia Sinica, No. 128 Academia Road, Section 2, Nankang Dist., 115, Taipei, Taiwan
| | - Chung-Yen Lin
- Institute of Information Science, Academia Sinica, No. 128 Academia Road, Section 2, Nankang Dist., 115, Taipei, Taiwan
| |
Collapse
|
27
|
Kim BJ, Scott DA. RERE deficiency causes retinal and optic nerve atrophy through degeneration of retinal cells. Dev Dyn 2021; 250:1398-1409. [PMID: 33742727 DOI: 10.1002/dvdy.330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 03/15/2021] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND The arginine-glutamic acid dipeptide repeats gene (RERE) encodes a nuclear receptor coregulator that modulates gene expression through its interaction with transcriptional machinery. In humans, RERE deficiency causes neurodevelopmental disorder with or without structural defects of the brain, eye, heart, and kidney (NEDBEH). Ophthalmological defects are seen in approximately one third of individuals with NEDBEH and in RERE-deficient mice which can serve as a useful animal model. RESULTS In mice, RERE is expressed in a subset of retinal ganglion cells (RGC), the lens epithelium, and the ciliary body during the embryonic period. RERE expression expands into the outer nuclear layer and the inner nuclear layer during the postnatal period. RERE-deficient mice have retinal and optic nerve atrophy. We show that RERE deficiency causes progressive loss of retinal cells and apoptosis of retinal cells in the ganglion cell layer as early as E17.5. The number of RGCs is also reduced in RERE-deficient embryos and mice. CONCLUSIONS We conclude that RERE is required to control the apoptosis of retinal cells in the developing retina, and that RERE deficiency results in the retina atrophy through degeneration of the retinal cells and optic nerve atrophy through the loss of RGCs.
Collapse
Affiliation(s)
- Bum Jun Kim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Daryl A Scott
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA.,Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
28
|
Raj B, Farrell JA, Liu J, El Kholtei J, Carte AN, Navajas Acedo J, Du LY, McKenna A, Relić Đ, Leslie JM, Schier AF. Emergence of Neuronal Diversity during Vertebrate Brain Development. Neuron 2020; 108:1058-1074.e6. [PMID: 33068532 PMCID: PMC8286448 DOI: 10.1016/j.neuron.2020.09.023] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 08/05/2020] [Accepted: 09/17/2020] [Indexed: 01/03/2023]
Abstract
Neurogenesis comprises many highly regulated processes including proliferation, differentiation, and maturation. However, the transcriptional landscapes underlying brain development are poorly characterized. We describe a developmental single-cell catalog of ∼220,000 zebrafish brain cells encompassing 12 stages from embryo to larva. We characterize known and novel gene markers for ∼800 clusters and provide an overview of the diversification of neurons and progenitors across these time points. We also introduce an optimized GESTALT lineage recorder that enables higher expression and recovery of Cas9-edited barcodes to query lineage segregation. Cell type characterization indicates that most embryonic neural progenitor states are transitory and transcriptionally distinct from neural progenitors of post-embryonic stages. Reconstruction of cell specification trajectories reveals that late-stage retinal neural progenitors transcriptionally overlap cell states observed in the embryo. The zebrafish brain development atlas provides a resource to define and manipulate specific subsets of neurons and to uncover the molecular mechanisms underlying vertebrate neurogenesis.
Collapse
Affiliation(s)
- Bushra Raj
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA.
| | - Jeffrey A Farrell
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Unit on Cell Specification and Differentiation, National Institute of Child Health and Human Development, NIH, Bethesda, MD 20814, USA
| | - Jialin Liu
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA; Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Jakob El Kholtei
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA; Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Adam N Carte
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Biozentrum, University of Basel, 4056 Basel, Switzerland; Systems, Synthetic, and Quantitative Biology Program, Harvard University, Cambridge, MA 02138, USA
| | - Joaquin Navajas Acedo
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA; Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Lucia Y Du
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA; Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Aaron McKenna
- Department of Molecular and Systems Biology, Dartmouth Geisel School of Medicine, Lebanon, NH 03756, USA
| | - Đorđe Relić
- Biozentrum, University of Basel, 4056 Basel, Switzerland; Swiss Institute of Bioinformatics (SIB), 4056 Basel, Switzerland
| | - Jessica M Leslie
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA
| | - Alexander F Schier
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA 02138, USA; Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA; Biozentrum, University of Basel, 4056 Basel, Switzerland; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA; Center for Brain Science, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
29
|
Fuhrmann JF, Buono L, Adelmann L, Martinez-Morales JR, Centanin L. Genetic developmental timing revealed by inter-species transplantations in fish. Development 2020; 147:dev.192500. [PMID: 33033120 DOI: 10.1242/dev.192500] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 10/02/2020] [Indexed: 12/22/2022]
Abstract
The path from a fertilised egg to an embryo involves the coordinated formation of cell types, tissues and organs. Developmental modules comprise discrete units specified by self-sufficient genetic programs that can interact with each other during embryogenesis. Here, we have taken advantage of the different span of embryonic development between two distantly related teleosts, zebrafish (Danio rerio) and medaka (Oryzias latipes) (3 and 9 days, respectively), to explore modularity principles. We report that inter-species blastula transplantations result in the ectopic formation of a retina formed by donor cells - a module. We show that the time taken for the retina to develop follows a genetic program: an ectopic zebrafish retina in medaka develops with zebrafish dynamics. Heterologous transplantation results in a temporal decoupling between the donor retina and host organism, illustrated by two paradigms that require retina-host interactions: lens recruitment and retino-tectal projections. Our results uncover a new experimental system for addressing temporal decoupling along embryonic development, and highlight the presence of largely autonomous but interconnected developmental modules that orchestrate organogenesis.
Collapse
Affiliation(s)
- Jana Franziska Fuhrmann
- Laboratory of Clonal Analysis, Center for Organismal Studies, Universität Heidelberg, INF230, 69120 Heidelberg, Germany
| | - Lorena Buono
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Carretera de Utrera km 1, 41013 Seville, Spain
| | - Leonie Adelmann
- Laboratory of Clonal Analysis, Center for Organismal Studies, Universität Heidelberg, INF230, 69120 Heidelberg, Germany
| | - Juan Ramón Martinez-Morales
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide, Carretera de Utrera km 1, 41013 Seville, Spain
| | - Lazaro Centanin
- Laboratory of Clonal Analysis, Center for Organismal Studies, Universität Heidelberg, INF230, 69120 Heidelberg, Germany
| |
Collapse
|
30
|
Piedade WP, Titialii-Torres K, Morris AC, Famulski JK. Proteasome-Mediated Regulation of Cdhr1a by Siah1 Modulates Photoreceptor Development and Survival in Zebrafish. Front Cell Dev Biol 2020; 8:594290. [PMID: 33330480 PMCID: PMC7719784 DOI: 10.3389/fcell.2020.594290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/22/2020] [Indexed: 01/05/2023] Open
Abstract
Congenital retinal dystrophies are a major cause of unpreventable and incurable blindness worldwide. Mutations in CDHR1, a retina specific cadherin, are associated with cone-rod dystrophy. The ubiquitin proteasome system (UPS) is responsible for mediating orderly and precise targeting of protein degradation to maintain biological homeostasis and coordinate proper development, including retinal development. Recently, our lab uncovered that the seven in absentia (Siah) family of E3 ubiquitin ligases play a role in optic fissure fusion and identified Cdhr1a as a potential target of Siah. Using two-color whole mount in situ hybridization and immunohistochemistry, we detected siah1 and cdhr1a co-expression as well as protein localization in the retinal outer nuclear layer (ONL), and more precisely in the connecting cilium of rods and cones between 3-5 days post fertilization (dpf). We confirmed that Siah1 targets Cdhr1a for proteasomal degradation by co-transfection and co-immunoprecipitation in cell culture. To analyze the functional importance of this interaction, we created two transgenic zebrafish lines that express siah1 or an inactive siah1 (siah1ΔRING) under the control of the heat shock promoter to modulate Siah activity during photoreceptor development. Overexpression of siah1, but not siah1ΔRING, resulted in a decrease in the number of rods and cones at 72 h post fertilization (hpf). The number of retinal ganglion cells, amacrine and bipolar cells was not affected by Siah1 overexpression, and there was no significant reduction of proliferating cells in the Siah1 overexpressing retina. We did, however, detect increased cell death, confirmed by an increase in the number of TUNEL + cells in the ONL, which was proteasome-dependent, as proteasome inhibition rescued the cell death phenotype. Furthermore, reduction in rods and cones resulting from increased Siah1 expression was rescued by injection of cdhr1a mRNA, and to an even greater extent by injection of a Siah1-insensitive cdhr1a variant mRNA. Lastly, CRISPR induced loss of Cdhr1a function phenocopied Siah1 overexpression resulting in a significant reduction of rods and cones. Taken together, our work provides the first evidence that Cdhr1a plays a role during early photoreceptor development and that Cdhr1a is regulated by Siah1 via the UPS.
Collapse
Affiliation(s)
| | | | | | - Jakub K. Famulski
- Department of Biology, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
31
|
Luz-Madrigal A, Grajales-Esquivel E, Tangeman J, Kosse S, Liu L, Wang K, Fausey A, Liang C, Tsonis PA, Del Rio-Tsonis K. DNA demethylation is a driver for chick retina regeneration. Epigenetics 2020; 15:998-1019. [PMID: 32290791 PMCID: PMC7518676 DOI: 10.1080/15592294.2020.1747742] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/26/2020] [Accepted: 03/09/2020] [Indexed: 01/01/2023] Open
Abstract
Cellular reprogramming resets the epigenetic landscape to drive shifts in transcriptional programmes and cell identity. The embryonic chick can regenerate a complete neural retina, after retinectomy, via retinal pigment epithelium (RPE) reprogramming in the presence of FGF2. In this study, we systematically analysed the reprogramming competent chick RPE prior to injury, and during different stages of reprogramming. In addition to changes in the expression of genes associated with epigenetic modifications during RPE reprogramming, we observed dynamic changes in histone marks associated with bivalent chromatin (H3K27me3/H3K4me3) and intermediates of the process of DNA demethylation including 5hmC and 5caC. Comprehensive analysis of the methylome by whole-genome bisulphite sequencing (WGBS) confirmed extensive rearrangements of DNA methylation patterns including differentially methylated regions (DMRs) found at promoters of genes associated with chromatin organization and fibroblast growth factor production. We also identified Tet methylcytosine dioxygenase 3 (TET3) as an important factor for DNA demethylation and retina regeneration, capable of reprogramming RPE in the absence of exogenous FGF2. In conclusion, we demonstrate that injury early in RPE reprogramming triggers genome-wide dynamic changes in chromatin, including bivalent chromatin and DNA methylation. In the presence of FGF2, these dynamic modifications are further sustained in the commitment to form a new retina. Our findings reveal active DNA demethylation as an important process that may be applied to remove the epigenetic barriers in order to regenerate retina in mammals. ABBREVIATIONS bp: Base pair; DMR: Differentially methylated region; DMC: Differentially methylated cytosines; GFP: Green fluorescent protein; PCR: Polymerase chain reaction. TET: Ten-eleven translocation; RPE: retinal pigment epithelium.
Collapse
Affiliation(s)
- Agustín Luz-Madrigal
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH, USA
- Department of Biology and Center for Stem Cell & Organoid Medicine (CuSTOM), Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Erika Grajales-Esquivel
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH, USA
| | - Jared Tangeman
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH, USA
| | - Sarah Kosse
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH, USA
| | - Lin Liu
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH, USA
| | - Kai Wang
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH, USA
| | - Andrew Fausey
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH, USA
| | - Chun Liang
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH, USA
- Department of Computer Science and Software Engineering, Miami University, Oxford, OH, USA
| | - Panagiotis A. Tsonis
- Department of Biology, University of Dayton and Center for Tissue Regeneration and Engineering at the University of Dayton (TREND), Dayton, OH, USA
| | - Katia Del Rio-Tsonis
- Department of Biology and Center for Visual Sciences at Miami University, Miami University, Oxford, OH, USA
| |
Collapse
|
32
|
Persiconi I, Cosmi F, Guadagno NA, Lupo G, De Stefano ME. Dystrophin Is Required for the Proper Timing in Retinal Histogenesis: A Thorough Investigation on the mdx Mouse Model of Duchenne Muscular Dystrophy. Front Neurosci 2020; 14:760. [PMID: 32982660 PMCID: PMC7487415 DOI: 10.3389/fnins.2020.00760] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/29/2020] [Indexed: 12/15/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal X-linked muscular disease caused by defective expression of the cytoskeletal protein dystrophin (Dp427). Selected autonomic and central neurons, including retinal neurons, express Dp427 and/or dystrophin shorter isoforms. Because of this, DMD patients may also experience different forms of cognitive impairment, neurological and autonomic disorders, and specific visual defects. DMD-related damages to the nervous system are established during development, suggesting a role for all dystrophin isoforms in neural circuit development and differentiation; however, to date, their function in retinogenesis has never been investigated. In this large-scale study, we analyzed whether the lack of Dp427 affects late retinogenesis in the mdx mouse, the most well studied animal model of DMD. Retinal gene expression and layer maturation, as well as neural cell proliferation, apoptosis, and differentiation, were evaluated in E18 and/or P0, P5, P10, and adult mice. In mdx mice, expression of Capn3, Id3 (E18-P5), and Dtnb (P5) genes, encoding proteins involved in different aspects of retina development and synaptogenesis (e.g., Calpain 3, DNA-binding protein inhibitor-3, and β-dystrobrevin, respectively), was transiently reduced compared to age-matched wild type mice. Concomitantly, a difference in the time required for the retinal ganglion cell layer to reach appropriate thickness was observed (P0–P5). Immunolabeling for specific cell markers also evidenced a significant dysregulation in the number of GABAergic amacrine cells (P5–P10), a transient decrease in the area immunopositive for the Vesicular Glutamate Transporter 1 (VGluT1) during ribbon synapse maturation (P10) and a reduction in the number of calretinin+ retinal ganglion cells (RGCs) (adults). Finally, the number of proliferating retinal progenitor cells (P5–P10) and apoptotic cells (P10) was reduced. These results support the hypothesis of a role for Dp427 during late retinogenesis different from those proposed in consolidated neural circuits. In particular, Dp427 may be involved in shaping specific steps of retina differentiation. Notably, although most of the above described quantitative alterations recover over time, the number of calretinin+ RGCs is reduced only in the mature retina. This suggests that alterations subtler than the timing of retinal maturation may occur, a hypothesis that demands further in-depth functional studies.
Collapse
Affiliation(s)
- Irene Persiconi
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Francesca Cosmi
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | | | - Giuseppe Lupo
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Maria Egle De Stefano
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy.,Center for Research in Neurobiology "Daniel Bovet", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
33
|
Gomes AL, Matos-Rodrigues GE, Frappart PO, Martins RAP. RINT1 Loss Impairs Retinogenesis Through TRP53-Mediated Apoptosis. Front Cell Dev Biol 2020; 8:711. [PMID: 32850831 PMCID: PMC7406574 DOI: 10.3389/fcell.2020.00711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/13/2020] [Indexed: 01/22/2023] Open
Abstract
Genomic instability in the central nervous system (CNS) is associated with defective neurodevelopment and neurodegeneration. Congenital human syndromes that affect the CNS development originate from mutations in genes of the DNA damage response (DDR) pathways. RINT1 (Rad50-interacting protein 1) is a partner of RAD50, that participates in the cellular responses to DNA double-strand breaks (DSB). Recently, we showed that Rint1 regulates cell survival in the developing brain and its loss led to premature lethality associated with genomic stability. To bypass the lethality of Rint1 inactivation in the embryonic brain and better understand the roles of RINT1 in CNS development, we conditionally inactivated Rint1 in retinal progenitor cells (RPCs) during embryogenesis. Rint1 loss led to accumulation of endogenous DNA damage, but RINT1 was not necessary for the cell cycle checkpoint activation in these neural progenitor cells. As a consequence, proliferating progenitors and postmitotic neurons underwent apoptosis causing defective neurogenesis of retinal ganglion cells, malformation of the optic nerve and blindness. Notably, inactivation of Trp53 prevented apoptosis of the RPCs and rescued the generation of retinal neurons and vision loss. Together, these results revealed an essential role for TRP53-mediated apoptosis in the malformations of the visual system caused by RINT1 loss and suggests that defective responses to DNA damage drive retinal malformations.
Collapse
Affiliation(s)
- Anielle L Gomes
- Programa de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabriel E Matos-Rodrigues
- Programa de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pierre-Olivier Frappart
- Institute of Toxicology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Rodrigo A P Martins
- Programa de Biologia Celular e do Desenvolvimento, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
34
|
Stanchfield ML, Webster SE, Webster MK, Linn CL. Involvement of HB-EGF/Ascl1/Lin28a Genes in Dedifferentiation of Adult Mammalian Müller Glia. Front Mol Biosci 2020; 7:200. [PMID: 32923455 PMCID: PMC7457012 DOI: 10.3389/fmolb.2020.00200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 07/24/2020] [Indexed: 12/14/2022] Open
Abstract
Previous studies from this lab have determined that dedifferentiation of Müller glia occurs after eye drop application of an α7 nicotinic acetylcholine receptor (nAChR) agonist, PNU-282987, to the adult rodent eye. PNU-282987 acts on α7 nAChRs on retinal pigment epithelial cells to stimulate production of Müller-derived progenitor cells (MDPCs) and ultimately lead to neurogenesis. This current study was designed to test the hypothesis that the activation of genes involved in the HB-EGF/Ascl1/Lin28a signaling pathway in Müller glia leads to the genesis of MDPCs. RNA-seq was performed on a Müller glial cell line (rMC-1) following contact with supernatant collected from a retinal pigment epithelial (RPE) cell line treated with PNU-282987. Differentially regulated genes were compared with published literature of Müller glia dedifferentiation that occurs in lower vertebrate regeneration and early mammalian development. HB-EGF was significantly up-regulated by 8 h and expression increased through 12 h. By 48 h, up-regulation of Ascl1 and Lin28a was observed, two genes known to be rapidly induced in dedifferentiating zebrafish Müller glia. Up-regulation of other genes known to be involved in mammalian development and zebrafish regeneration were also observed, as well as down-regulation of some factors necessary for Müller glia cell identity. RNA-seq results were verified using qRT-PCR. Using immunocytochemistry, the presence of markers associated with MDCP identity, Otx2, Nestin, and Vsx2, were found to be expressed in the 48 h treatment group cultures. This study is novel in its demonstration that Müller glia in adult rodents can be induced into regenerative activity by stimulating genes involved in the HB-EGF/Ascl1/Lin28a pathway that leads to MDPCs after introducing conditioned media from PNU-282987 treated RPE. This study furthers our understanding of the mechanism by which Müller glia dedifferentiate in response to PNU-282987 in the adult mammalian retina.
Collapse
Affiliation(s)
| | | | | | - Cindy L. Linn
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, United States
| |
Collapse
|
35
|
Lu Y, Shiau F, Yi W, Lu S, Wu Q, Pearson JD, Kallman A, Zhong S, Hoang T, Zuo Z, Zhao F, Zhang M, Tsai N, Zhuo Y, He S, Zhang J, Stein-O'Brien GL, Sherman TD, Duan X, Fertig EJ, Goff LA, Zack DJ, Handa JT, Xue T, Bremner R, Blackshaw S, Wang X, Clark BS. Single-Cell Analysis of Human Retina Identifies Evolutionarily Conserved and Species-Specific Mechanisms Controlling Development. Dev Cell 2020; 53:473-491.e9. [PMID: 32386599 PMCID: PMC8015270 DOI: 10.1016/j.devcel.2020.04.009] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/05/2020] [Accepted: 04/10/2020] [Indexed: 01/08/2023]
Abstract
The development of single-cell RNA sequencing (scRNA-seq) has allowed high-resolution analysis of cell-type diversity and transcriptional networks controlling cell-fate specification. To identify the transcriptional networks governing human retinal development, we performed scRNA-seq analysis on 16 time points from developing retina as well as four early stages of retinal organoid differentiation. We identified evolutionarily conserved patterns of gene expression during retinal progenitor maturation and specification of all seven major retinal cell types. Furthermore, we identified gene-expression differences between developing macula and periphery and between distinct populations of horizontal cells. We also identified species-specific patterns of gene expression during human and mouse retinal development. Finally, we identified an unexpected role for ATOH7 expression in regulation of photoreceptor specification during late retinogenesis. These results provide a roadmap to future studies of human retinal development and may help guide the design of cell-based therapies for treating retinal dystrophies.
Collapse
Affiliation(s)
- Yufeng Lu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fion Shiau
- John F. Hardesty, MD, Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wenyang Yi
- Hefei National Laboratory for Physical Sciences, at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China
| | - Suying Lu
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health Systems, Department of Ophthalmology and Vision Science, and Department of Lab Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Qian Wu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Joel D Pearson
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health Systems, Department of Ophthalmology and Vision Science, and Department of Lab Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Alyssa Kallman
- Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Suijuan Zhong
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Thanh Hoang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Zhentao Zuo
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Fangqi Zhao
- Obstetrics and Gynecology Medical Center of Severe Cardiovascular of Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Mei Zhang
- Hefei National Laboratory for Physical Sciences, at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Nicole Tsai
- Departments of Ophthalmology and Physiology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Yan Zhuo
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Sheng He
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Jun Zhang
- Obstetrics and Gynecology Medical Center of Severe Cardiovascular of Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Genevieve L Stein-O'Brien
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Thomas D Sherman
- Department of Oncology, Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xin Duan
- Departments of Ophthalmology and Physiology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Elana J Fertig
- Department of Oncology, Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Data Intensive Engineering and Science, Johns Hopkins University, Baltimore, MD 21218, USA; Institute for Computational Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Mathematical Institute for Data Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Loyal A Goff
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Donald J Zack
- Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - James T Handa
- Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tian Xue
- Hefei National Laboratory for Physical Sciences, at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, School of Life Sciences, University of Science and Technology of China, Hefei 230026, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China.
| | - Rod Bremner
- Lunenfeld Tanenbaum Research Institute, Mt Sinai Hospital, Sinai Health Systems, Department of Ophthalmology and Vision Science, and Department of Lab Medicine and Pathobiology, University of Toronto, Toronto, ON M5G 1X5, Canada.
| | - Seth Blackshaw
- Department of Ophthalmology, Wilmer Eye Institute Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Human Systems Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | - Xiaoqun Wang
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Brain Disorders, Beijing 100069, China.
| | - Brian S Clark
- John F. Hardesty, MD, Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
36
|
Wu M, Liu J, Li F, Huang S, He J, Xue Y, Fu T, Feng S, Li Z. Antibiotic-induced dysbiosis of gut microbiota impairs corneal development in postnatal mice by affecting CCR2 negative macrophage distribution. Mucosal Immunol 2020; 13:47-63. [PMID: 31434991 PMCID: PMC6914671 DOI: 10.1038/s41385-019-0193-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 06/18/2019] [Accepted: 07/17/2019] [Indexed: 02/04/2023]
Abstract
Antibiotics are extremely useful, but they can cause adverse impacts on host bodies. We found that antibiotic treatment altered the composition of the gut microbiota and the gene expression profile in the corneal tissues of postnatal mice and decreased the corneal size and thickness, the angiogenesis of limbal blood vessels, and the neurogenesis of corneal nerve fibers. The reconstitution of the gut microbiota with fecal transplants in antibiotic-treated mice largely reversed these impairments in corneal development. Furthermore, C-C chemokine receptor type 2 negative (CCR2-) macrophages were confirmed to participate in corneal development, and their distribution in the cornea was regulated by the gut microbiota. We propose that the CCR2- macrophage population is a crucial mediator through which gut microbiota affect corneal development in postnatal mice. In addition, probiotics were shown to have the potential effect of restoring corneal development in antibiotic-treated mice. Abx-induced gut dysbiosis has significant, long-term effects on the development of the cornea, and reversal of these suppressive effects takes a long time.
Collapse
Affiliation(s)
- Mingjuan Wu
- 0000 0004 1790 3548grid.258164.cInternational Ocular Surface Research Center, Institute of Ophthalmology, and Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China
| | - Jun Liu
- 0000 0004 1790 3548grid.258164.cInternational Ocular Surface Research Center, Institute of Ophthalmology, and Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China
| | - Fanying Li
- 0000 0004 1790 3548grid.258164.cInternational Ocular Surface Research Center, Institute of Ophthalmology, and Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China ,0000 0004 1790 3548grid.258164.cDepartment of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, China
| | - Shuoya Huang
- 0000 0004 1790 3548grid.258164.cInternational Ocular Surface Research Center, Institute of Ophthalmology, and Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China ,0000 0004 1760 3828grid.412601.0Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jingxin He
- 0000 0004 1790 3548grid.258164.cInternational Ocular Surface Research Center, Institute of Ophthalmology, and Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China ,0000 0004 1760 3828grid.412601.0Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Yunxia Xue
- 0000 0004 1790 3548grid.258164.cInternational Ocular Surface Research Center, Institute of Ophthalmology, and Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China
| | - Ting Fu
- 0000 0004 1790 3548grid.258164.cInternational Ocular Surface Research Center, Institute of Ophthalmology, and Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China
| | - Shanshan Feng
- 0000 0004 1790 3548grid.258164.cInternational Ocular Surface Research Center, Institute of Ophthalmology, and Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China
| | - Zhijie Li
- 0000 0004 1790 3548grid.258164.cInternational Ocular Surface Research Center, Institute of Ophthalmology, and Key Laboratory for Regenerative Medicine, Jinan University, Guangzhou, China ,0000 0004 1760 3828grid.412601.0Department of Ophthalmology, The First Affiliated Hospital of Jinan University, Guangzhou, China ,grid.414011.1Department of Ophthalmology, Henan Provincial People’s Hospital, Zhengzhou, China
| |
Collapse
|
37
|
Sharma S, Chakravarthy H, Suresh G, Devanathan V. Adult Goat Retinal Neuronal Culture: Applications in Modeling Hyperglycemia. Front Neurosci 2019; 13:983. [PMID: 31607843 PMCID: PMC6756134 DOI: 10.3389/fnins.2019.00983] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 09/02/2019] [Indexed: 12/13/2022] Open
Abstract
Culture of adult neurons of the central nervous system (CNS) can provide a unique model system to explore neurodegenerative diseases. The CNS includes neurons and glia of the brain, spinal cord and retina. Neurons in the retina have the advantage of being the most accessible cells of the CNS, and can serve as a reliable mirror to the brain. Typically, primary cultures utilize fetal rodent neurons, but very rarely adult neurons from larger mammals. Here, we cultured primary retinal neurons isolated from adult goat up to 10 days, and established an in vitro model of hyperglycemia for performing morphological and molecular characterization studies. Immunofluorescence staining revealed that approximately 30–40% of cultured cells expressed neuronal markers. Next, we examined the relative expression of cell adhesion molecules (CAMs) in adult goat brain and retina. We also studied the effect of different glucose concentrations and media composition on the growth and expression of CAMs in cultured retinal neurons. Hyperglycemia significantly enhances neurite outgrowth in adult retinal neurons in culture. Expression of CAMs such as Caspr1, Contactin1 and Prion is downregulated in the presence of high glucose. Hyperglycemia downregulates the expression of the transcription factor CCAAT/enhancer binding protein (C/EBP α), predicted to bind CAM gene promoters. Collectively, our study demonstrates that metabolic environment markedly affects transcriptional regulation of CAMs in adult retinal neurons in culture. The effect of hyperglycemia on CAM interactions, as well as related changes in intracellular signaling pathways in adult retinal neurons warrants further investigation.
Collapse
Affiliation(s)
- Sapana Sharma
- Department of Biology, Indian Institute of Science Education and Research (IISER), Tirupati, India
| | - Harshini Chakravarthy
- Department of Biology, Indian Institute of Science Education and Research (IISER), Tirupati, India
| | - Gowthaman Suresh
- Department of Biology, Indian Institute of Science Education and Research (IISER), Tirupati, India
| | - Vasudharani Devanathan
- Department of Biology, Indian Institute of Science Education and Research (IISER), Tirupati, India
| |
Collapse
|
38
|
Xia X, Teotia P, Ahmad I. miR-29c regulates neurogliogenesis in the mammalian retina through REST. Dev Biol 2019; 450:90-100. [PMID: 30914322 DOI: 10.1016/j.ydbio.2019.03.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/05/2019] [Accepted: 03/20/2019] [Indexed: 10/27/2022]
Abstract
In the developing central nervous system, including its simple and accessible model retina, neurogenesis is followed by gliogenesis. However, the mechanism underlying the neurogliogenic switch remains poorly understood despite the identification of several regulatory genes, associated with the lineage identity and transition. The mechanism may involve cross talks between regulatory genes, facilitated through microRNAs. Here, we posit miR-29c as one of the regulatory miRNAs that may influence neuronal versus glial differentiation. We observed that the temporal patterns of miR-29c expression corresponded with late retinal histogenesis, the stage in the developing retina when neurogliogenic decision predominantly occurs. Examination of the effects of miR-29c on neurogliogenesis by the perturbation of function approach revealed that miR-29c preferentially facilitated differentiation of late RPCs into rod photoreceptors and bipolar cells, the late-born neurons, at the expense of Müller glia, the sole glia generated by retinal progenitor cells. We further observed that miR-29c facilitated neurogenesis and inhibited gliogenesis by regulating the expression of RE-1 silencing transcription factor (REST), which encodes a transcriptional repressor of cell cycle regulators and neuronal genes. Thus, miR-29c may influence neurogliogenic decision in the developing retina by regulating the instructive out put of a molecular axis helmed by REST.
Collapse
Affiliation(s)
- Xiaohuan Xia
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Pooja Teotia
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Iqbal Ahmad
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
39
|
Pluripotent Stem Cells as Models of Retina Development. Mol Neurobiol 2019; 56:6056-6070. [DOI: 10.1007/s12035-019-1504-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 01/21/2019] [Indexed: 01/01/2023]
|
40
|
Taylor SM, Giuffre E, Moseley P, Hitchcock PF. The MicroRNA, miR-18a, Regulates NeuroD and Photoreceptor Differentiation in the Retina of Zebrafish. Dev Neurobiol 2019; 79:202-219. [PMID: 30615274 PMCID: PMC6351175 DOI: 10.1002/dneu.22666] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/21/2022]
Abstract
During embryonic retinal development, six types of retinal neurons are generated from multipotent progenitors in a strict spatiotemporal pattern. This pattern requires cell cycle exit (i.e. neurogenesis) and differentiation to be precisely regulated in a lineage‐specific manner. In zebrafish, the bHLH transcription factor NeuroD governs photoreceptor genesis through Notch signaling but also governs photoreceptor differentiation though distinct mechanisms that are currently unknown. Also unknown are the mechanisms that regulate NeuroD and the spatiotemporal pattern of photoreceptor development. Members of the miR‐17‐92 microRNA cluster regulate CNS neurogenesis, and a member of this cluster, miR‐18a, is predicted to target neuroD mRNA. The purpose of this study was to determine if, in the developing zebrafish retina, miR‐18a regulates NeuroD and if it plays a role in photoreceptor development. Quantitative RT‐PCR showed that, of the three miR‐18 family members (miR‐18a, b, and c), miR‐18a expression most closely parallels neuroD expression. Morpholino oligonucleotides and CRISPR/Cas9 gene editing were used for miR‐18a loss‐of‐function (LOF) and both resulted in larvae with more mature photoreceptors at 70 hpf without affecting cell proliferation. Western blot showed that miR‐18a LOF increases NeuroD protein levels and in vitro dual luciferase assay showed that miR‐18a directly interacts with the 3′ UTR of neuroD. Finally, tgif1 mutants have increased miR‐18a expression, less NeuroD protein and fewer mature photoreceptors, and the photoreceptor deficiency is rescued by miR‐18a knockdown. Together, these results show that, independent of neurogenesis, miR‐18a regulates the timing of photoreceptor differentiation and indicate that this occurs through post‐transcriptional regulation of NeuroD.
Collapse
Affiliation(s)
- Scott M Taylor
- Department of Biology, University of West Florida, 11000 University Parkway, Pensacola, Florida, 32514
| | - Emily Giuffre
- Department of Biology, University of West Florida, 11000 University Parkway, Pensacola, Florida, 32514
| | - Patience Moseley
- Department of Biology, University of West Florida, 11000 University Parkway, Pensacola, Florida, 32514
| | - Peter F Hitchcock
- Ophthalmology and Visual Sciences, University of Michigan, W. K. Kellogg Eye Center, 1000 Wall Street, Ann Arbor, Michigan, 48105
| |
Collapse
|
41
|
Surendran H, Rathod RJ, Pal R. Generation of Transplantable Retinal Pigmented Epithelial (RPE) Cells for Treatment of Age-Related Macular Degeneration (AMD). Methods Mol Biol 2019; 2045:283-298. [PMID: 29896658 DOI: 10.1007/7651_2018_140] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Age-related macular degeneration (AMD) is the foremost cause of blindness in people over the age of 60 worldwide. Clinically, this disease starts with distortion in central vision eventually leading to legal blindness. Vision loss has a significant impact on quality of life and incurs a substantial cost to the economy. Furthermore, AMD is a complex and progressive neurodegenerative disorder that triggers visual impairment due to the loss of retinal pigmented epithelium (RPE) and the light-sensitive photoreceptors that they support, protect and provide nutrition. Currently, there is no curative treatment for the most common form of this disease, i.e., dry AMD. A novel approach to treat AMD involves the transplantation of RPE cells derived from human induced pluripotent stem cells (iPSCs) in the outer retina. These iPSC-derived RPE cells not only show characteristics similar to native RPE but also could replace as well as regenerate damaged pathologic RPE and produce supportive growth factors and cytokines. Several clinical trials are being conducted taking advantage of a variety of cell- and tissue engineering-based approaches. Here, we present a simple, cost effective, and scalable cell-culture model for generation of purified RPE thus providing the foundation for developing an allogeneic cell therapy for AMD.
Collapse
Affiliation(s)
- Harshini Surendran
- Eyestem Research, Centre for Cellular and Molecular Platforms (CCAMP), Bangalore, India
| | - Reena J Rathod
- Eyestem Research, Centre for Cellular and Molecular Platforms (CCAMP), Bangalore, India
| | - Rajarshi Pal
- Eyestem Research, Centre for Cellular and Molecular Platforms (CCAMP), Bangalore, India.
| |
Collapse
|
42
|
Hunt NC, Hallam D, Chichagova V, Steel DH, Lako M. The Application of Biomaterials to Tissue Engineering Neural Retina and Retinal Pigment Epithelium. Adv Healthc Mater 2018; 7:e1800226. [PMID: 30175520 DOI: 10.1002/adhm.201800226] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/16/2018] [Indexed: 12/21/2022]
Abstract
The prevalence of degenerative retinal disease is ever increasing as life expectancy rises globally. The human retina fails to regenerate and the use of human embryonic stem cells (hESCs) and human-induced pluripotent stem cells (hiPSCs) to engineer retinal tissue is of particular interest due to the limited availability of suitable allogeneic or autologous tissue. Retinal tissue and its development are well characterized, which have resulted in robust assays to assess the development of tissue-engineered retina. Retinal tissue can be generated in vitro from hESCs and hiPSCs without biomaterial scaffolds, but despite advancements, protocols remain slow, expensive, and fail to result in mature functional tissue. Several recent studies have demonstrated the potential of biomaterial scaffolds to enhance generation of hESC/hiPSC-derived retinal tissue, including synthetic polymers, silk, alginate, hyaluronic acid, and extracellular matrix molecules. This review outlines the advances that have been made toward tissue-engineered neural retina and retinal pigment epithelium (RPE) for clinical application in recent years, including the success of clinical trials involving transplantation of cells and tissue to promote retinal repair; and the evidence from in vitro and animal studies that biomaterials can enhance development and integration of retinal tissue.
Collapse
Affiliation(s)
- Nicola C. Hunt
- Newcastle UniversityInstitute of Genetic MedicineInternational Centre for Life Central Parkway Newcastle NE1 3BZ UK
| | - Dean Hallam
- Newcastle UniversityInstitute of Genetic MedicineInternational Centre for Life Central Parkway Newcastle NE1 3BZ UK
| | - Valeria Chichagova
- Newcastle UniversityInstitute of Genetic MedicineInternational Centre for Life Central Parkway Newcastle NE1 3BZ UK
- Biomedicine WestInternational Centre for LifeTimes SquareNewcastle upon Tyne NE1 4EP UK
| | - David H. Steel
- Newcastle UniversityInstitute of Genetic MedicineInternational Centre for Life Central Parkway Newcastle NE1 3BZ UK
| | - Majlinda Lako
- Newcastle UniversityInstitute of Genetic MedicineInternational Centre for Life Central Parkway Newcastle NE1 3BZ UK
| |
Collapse
|
43
|
Laboissonniere LA, Smith CL, Mesenbrink J, Chowdhury R, Burney A, Lang M, Sierra M, Stark A, Maldonado-Casalduc G, Muller M, Trimarchi JM. ALS-associated genes display CNS expression in the developing zebrafish. Gene Expr Patterns 2018; 30:14-31. [DOI: 10.1016/j.gep.2018.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 08/03/2018] [Accepted: 08/17/2018] [Indexed: 12/11/2022]
|
44
|
Kovács-Valasek A, Szalontai B, Sétáló G, Gábriel R. Sensitive fluorescent hybridisation protocol development for simultaneous detection of microRNA and cellular marker proteins (in the retina). Histochem Cell Biol 2018; 150:557-566. [PMID: 30088096 PMCID: PMC6182695 DOI: 10.1007/s00418-018-1705-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2018] [Indexed: 01/20/2023]
Abstract
Nowadays, increasing number of microRNAs are found to have crucial roles in various physiological processes through gene expression regulation via RNA silencing as a result of base pairing with complementary mRNA sequences. To reveal the spatial distribution of microRNA expression in tissues, in situ hybridisation is the only method developed to date. This work aims to provide a novel approach to obtain information on the possible involvement of microRNA-s in regulatory processes under experimental conditions by enhancing fluorescent detection of microRNA labelling. Developing Wistar rats were used as a model system to analyse retinal microRNA expression in the first 3 postnatal weeks. Using cryosections, the crucial elements of optimal labels were (1) the concentration and duration of proteinase K treatment, (2) hybridisation temperature of microRNA probes and (3) temperature of stringency washes. Further improvements made possible to combine our in situ hybridisation protocol with double-label immunofluorescence allowing for the simultaneous detection of microRNA-s with high sensitivity and a neuronal cell marker and/or a synaptic marker protein. Thus, the regulatory microRNA-s can be localised in an identified cell type along with its potential target protein. We believe that our protocol can be easily adapted for a variety of tissues of different origins, developmental stages and experimental conditions.
Collapse
Affiliation(s)
- Andrea Kovács-Valasek
- Department of Experimental Zoology and Neurobiology, University of Pécs, Ifjúság útja 6, Pécs, 7624, Hungary
| | - Bálint Szalontai
- János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - György Sétáló
- Department of Medical Biology, University of Pécs, Pécs, Hungary
| | - Robert Gábriel
- Department of Experimental Zoology and Neurobiology, University of Pécs, Ifjúság útja 6, Pécs, 7624, Hungary.
- János Szentágothai Research Centre, University of Pécs, Pécs, Hungary.
| |
Collapse
|
45
|
Maurer KA, Kowalchuk A, Shoja-Taheri F, Brown NL. Integral bHLH factor regulation of cell cycle exit and RGC differentiation. Dev Dyn 2018; 247:965-975. [PMID: 29770538 PMCID: PMC6105502 DOI: 10.1002/dvdy.24638] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/05/2018] [Accepted: 05/05/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND In the developing mouse embryo, the bHLH transcription factor Neurog2 is transiently expressed by retinal progenitor cells and required for the initial wave of neurogenesis. Remarkably, another bHLH factor, Ascl1, normally not present in the embryonic Neurog2 retinal lineage, can rescue the temporal phenotypes of Neurog2 mutants. RESULTS Here we show that Neurog2 simultaneously promotes terminal cell cycle exit and retinal ganglion cell differentiation, using mitotic window labeling and integrating these results with retinal marker quantifications. We also analyzed the transcriptomes of E12.5 GFP-expressing cells from Neurog2GFP/+ , Neurog2GFP/GFP , and Neurog2Ascl1KI/GFP eyes, and validated the most significantly affected genes using qPCR assays. CONCLUSIONS Our data support the hypothesis that Neurog2 acts at the top of a retinal bHLH transcription factor hierarchy. The combined expression levels of these downstream factors are sufficiently induced by ectopic Ascl1 to restore RGC genesis, highlighting the robustness of this gene network during retinal ganglion cell neurogenesis. Developmental Dynamics 247:965-975, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Kate A. Maurer
- Division of Developmental Biology, Cincinnati Children’s Hospital Research Foundation, Cincinnati, OH, 45229
| | - Angelica Kowalchuk
- Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Davis, CA 95616
| | - Farnaz Shoja-Taheri
- Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Davis, CA 95616
| | - Nadean L. Brown
- Division of Developmental Biology, Cincinnati Children’s Hospital Research Foundation, Cincinnati, OH, 45229
- Department of Cell Biology and Human Anatomy, University of California Davis, School of Medicine, Davis, CA 95616
| |
Collapse
|
46
|
Pogue AI, Lukiw WJ. Up-regulated Pro-inflammatory MicroRNAs (miRNAs) in Alzheimer's disease (AD) and Age-Related Macular Degeneration (AMD). Cell Mol Neurobiol 2018; 38:1021-1031. [PMID: 29302837 PMCID: PMC11481951 DOI: 10.1007/s10571-017-0572-3] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 12/21/2017] [Indexed: 11/25/2022]
Abstract
Alzheimer's disease (AD) of the brain neocortex and age-related macular degeneration (AMD) of the retina are two complex neurodegenerative disorders, which (i) involve the progressive dysregulation and deterioration of multiple neurobiological signaling pathways, (ii) exhibit the temporal accumulation of pro-inflammatory lesions including the amyloid beta (Aβ) peptide-containing senile plaques of AD and the drusen of AMD, and (iii) culminate in an insidious inflammatory neurodegeneration ending, respectively, in neural cell atrophy and death and progressive loss of cognition and central visual function. Recent independent research studies have indicated that AD and AMD share common, pathological signaling defects and disease mechanisms at the molecular genetic level. Using high-integrity total RNA samples pooled from AD brain and AMD retina, microfluidic hybridization miRNA arrays, and bioinformatics, the current study was undertaken to quantify microRNA (miRNA) speciation and complexity common to both AD and AMD. These small non-coding (sncRNAs) are known to post-transcriptionally regulate multiple neurobiological pathways and an abundance of research information has already been generated on the roles of these miRNAs in pathological situations involving inflammatory neuropathology and neural cell decline. Here, for the first time, we report the sequence and abundance of a septet of sncRNAs including miRNA-7, miRNA-9-1, miRNA-23a/miRNA-27a, miRNA-34a, miRNA-125b-1, miRNA-146a, and miRNA-155 that are significantly increased in abundance and common to both AD-affected superior temporal lobe neocortex (Brodmann A22) and the AMD-affected macular region of the retina. Bioinformatics, miRNA-mRNA complementarity, next-gen RNA sequencing, and feature alignment analysis further indicate that these 7 up-regulated miRNAs have the potential to interact with and down-regulate ~ 9460 target messenger RNAs (mRNAs; about 3.5% of the genome) involved in the synchronization of amyloid production and clearance, phagocytosis, innate-immune, pro-inflammatory, and neurotrophic signaling and/or synaptogenesis in diseased tissues.
Collapse
Affiliation(s)
| | - Walter J Lukiw
- Alchem Biotech Research, Toronto, ON, Canada.
- Neuroscience Center, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112-2272, USA.
- Department of Neurology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112-2272, USA.
- Department of Ophthalmology, Louisiana State University School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, 70112-2272, USA.
- LSU Neuroscience Center, Louisiana State University Health Sciences Center, 2020 Gravier Street, Suite 904, New Orleans, LA, 70112-2272, USA.
| |
Collapse
|
47
|
Ashok A, Karmakar S, Chandel R, Ravikumar R, Dalal S, Kong Q, Singh N. Prion protein modulates iron transport in the anterior segment: Implications for ocular iron homeostasis and prion transmission. Exp Eye Res 2018; 175:1-13. [PMID: 29859760 PMCID: PMC6167182 DOI: 10.1016/j.exer.2018.05.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 05/17/2018] [Accepted: 05/30/2018] [Indexed: 12/14/2022]
Abstract
Iron is an essential biometal in the aqueous humor, the principal source of nutrients for the avascular cornea and the lens. Here, we explored whether the ciliary body (CB), the source of aqueous humor, transports iron, and if the prion protein (PrPC) facilitates this process as in the outer retina. Using a combination of human, bovine, and mouse eyes as models, we report the expression of iron export proteins ferroportin and ceruloplasmin, and major iron uptake and storage proteins transferrin, transferrin receptor, and ferritin in the ciliary epithelium, indicating active exchange of iron at this site. Ferroportin and transferrin receptor are also expressed in the corneal endothelium. However, the relative expression of iron export and uptake proteins suggests export from the ciliary epithelium and import by corneal endothelium. In addition, abundant expression of PrPC, a ferrireductase that facilitates iron transport, is noted in pigmented and non-pigmented epithelium of the CB, posterior pigmented epithelium of the iris, corneal endothelium and epithelium, and lens epithelium. Notably, majority of PrPC in the ciliary epithelium is cleaved at the β-site as in retinal pigment epithelial cells, suggesting a role in iron transport. Most of the PrPC in the cornea, however, is full-length, and susceptible to aggregation by intracerebrally inoculated PrP-scrapie, an infectious conformation of PrPC responsible for human and animal prion disorders. Soluble PrPC is present in the aqueous and vitreous humor, a provocative observation with significant implications. Together, these observations suggest independent cycling of iron in the anterior segment, and a prominent role of PrPC in this process. Aggregation of PrPC in the cornea of PrP-scrapie-infected animals raises the alarming possibility of transmission of animal prions through corneal abrasions.
Collapse
Affiliation(s)
- Ajay Ashok
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Shilpita Karmakar
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Rajeev Chandel
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Ranjana Ravikumar
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Stuti Dalal
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Qingzhong Kong
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Neena Singh
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
48
|
Álvarez-Hernán G, Sánchez-Resino E, Hernández-Núñez I, Marzal A, Rodríguez-León J, Martín-Partido G, Francisco-Morcillo J. Retinal histogenesis in an altricial avian species, the zebra finch (Taeniopygia guttata, Vieillot 1817). J Anat 2018; 233:106-120. [PMID: 29582431 DOI: 10.1111/joa.12809] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2018] [Indexed: 01/11/2023] Open
Abstract
Comparative developmental studies have shown that the retina of altricial fish and mammals is incompletely developed at birth, and that, during the first days of life, maturation proceeds rapidly. In contrast, precocial fish and mammals are born with fully differentiated retinas. Concerning birds, knowledge about retinal development is generally restricted to a single order of precocial birds, Galliformes, due to the fact that both the chicken and the Japanese quail are considered model systems. However, comparison of embryonic pre-hatchling retinal development between altricial and precocial birds has been poorly explored. The purpose of this study was to examine the morphogenesis and histogenesis of the retina in the altricial zebra finch (Taeniopygia guttata, Vieillot 1817) and compare the results with those from previous studies in the precocial chicken. Several maturational features (morphogenesis of the optic vesicle and optic cup, appearance of the first differentiated neurons, the period in which the non-apical cell divisions are observable, and the emergence of the plexiform layers) were found to occur at later stages in the zebra finch than in the chicken. At hatching, the retina of T. guttata showed the typical cytoarchitecture of the mature tissue, although features of immaturity were still observable, such as a ganglion cell layer containing many thick cells, very thin plexiform layers, and poorly developed photoreceptors. Moreover, abundant mitotic activity was detected in the entire retina, even in the regions where the layering was complete. The circumferential marginal zone was very prominent and showed abundant mitotic activity. The partially undifferentiated stage of maturation at hatching makes the T. guttata retina an appropriate model with which to study avian postnatal retinal neurogenesis.
Collapse
Affiliation(s)
- Guadalupe Álvarez-Hernán
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Elena Sánchez-Resino
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Ismael Hernández-Núñez
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Alfonso Marzal
- Área de Zoología, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Joaquín Rodríguez-León
- Área de Anatomía Humana, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Medicina, Universidad de Extremadura, Badajoz, Spain
| | - Gervasio Martín-Partido
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| | - Javier Francisco-Morcillo
- Área de Biología Celular, Departamento de Anatomía, Biología Celular y Zoología, Facultad de Ciencias, Universidad de Extremadura, Badajoz, Spain
| |
Collapse
|
49
|
de Almeida-Pereira L, Repossi MG, Magalhães CF, Azevedo RDF, Corrêa-Velloso JDC, Ulrich H, Ventura ALM, Fragel-Madeira L. P2Y 12 but not P2Y 13 Purinergic Receptor Controls Postnatal Rat Retinogenesis In Vivo. Mol Neurobiol 2018; 55:8612-8624. [PMID: 29574630 DOI: 10.1007/s12035-018-1012-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 03/16/2018] [Indexed: 12/16/2022]
Abstract
Adenine nucleotides through P2Y1 receptor stimulation are known to control retinal progenitor cell (RPC) proliferation by modulating expression of the p57KIP2, a cell cycle regulator. However, the role of Gi protein-coupled P2Y12 and P2Y13 receptors also activated by adenine nucleotides in RPC proliferation is still unknown. Gene expression of the purinergic P2Y12 subtype was detected in rat retina during early postnatal days (P0 to P5), while expression levels of P2Y13 were low. Immunohistochemistry assays performed with rat retina on P3 revealed P2Y12 receptor expression in both Ki-67-positive cells in the neuroblastic layer and Ki-67-negative cells in the ganglion cell layer and inner nuclear layer. Nonetheless, P2Y13 receptor expression could not be detected in any stratum of rat retina. Intravitreal injection of PSB 0739 or clopidogrel, both selective P2Y12 receptor antagonists, increased by 20 and 15%, respectively, the number of Ki-67-positive cells following 24 h of exposure. Moreover, the P2Y12 receptor inhibition increased cyclin D1 and decreased p57KIP2 expression. However, there were no changes in the S phase of the cell cycle (BrdU-positive cells) or in mitosis (phospho-histone-H3-positive cells). Interestingly, an increase in the number of cyclin D1/TUNEL-positive cells after treatment with PSB 0739 was observed. These data suggest that activation of P2Y12 receptors is required for the successful exit of RPCs from cell cycle in the postnatal rat retina.
Collapse
Affiliation(s)
- Luana de Almeida-Pereira
- Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - Marinna Garcia Repossi
- Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - Camila Feitosa Magalhães
- Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | | | | | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | | - Lucianne Fragel-Madeira
- Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói, Brazil.
| |
Collapse
|
50
|
Rathod R, Surendran H, Battu R, Desai J, Pal R. Induced pluripotent stem cells (iPSC)-derived retinal cells in disease modeling and regenerative medicine. J Chem Neuroanat 2018; 95:81-88. [PMID: 29448001 DOI: 10.1016/j.jchemneu.2018.02.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 02/02/2018] [Accepted: 02/10/2018] [Indexed: 12/19/2022]
Abstract
Retinal degenerative disorders are a leading cause of the inherited, irreversible and incurable vision loss. While various rodent model systems have provided crucial information in this direction, lack of disease-relevant tissue availability and species-specific differences have proven to be a major roadblock. Human induced pluripotent stem cells (iPSC) have opened up a whole new avenue of possibilities not just in understanding the disease mechanism but also potential therapeutic approaches towards a cure. In this review, we have summarized recent advances in the methods of deriving retinal cell types from iPSCs which can serve as a renewable source of disease-relevant cell population for basic as well as translational studies. We also provide an overview of the ongoing efforts towards developing a suitable in vitro model for modeling retinal degenerative diseases. This basic understanding in turn has contributed to advances in translational goals such as drug screening and cell-replacement therapies. Furthermore we discuss gene editing approaches for autologous repair of genetic disorders and allogeneic transplantation of stem cell-based retinal derivatives for degenerative disorders with an ultimate goal to restore vision. It is pertinent to note however, that these exciting new developments throw up several challenges that need to be overcome before their full clinical potential can be realized.
Collapse
Affiliation(s)
- Reena Rathod
- Eyestem Research Private Limited, Centre for Cellular and Molecular Platforms (CCAMP), National Centre for Biological Sciences-Tata Institute of Fundamental Research (NCBS-TIFR), GKVK Campus, Bangalore, 560065, India
| | - Harshini Surendran
- Eyestem Research Private Limited, Centre for Cellular and Molecular Platforms (CCAMP), National Centre for Biological Sciences-Tata Institute of Fundamental Research (NCBS-TIFR), GKVK Campus, Bangalore, 560065, India
| | - Rajani Battu
- Eyestem Research Private Limited, Centre for Cellular and Molecular Platforms (CCAMP), National Centre for Biological Sciences-Tata Institute of Fundamental Research (NCBS-TIFR), GKVK Campus, Bangalore, 560065, India; Centre for Eye Genetics and Research, Cytecare Hospital, Bellary Road, Bangalore, 560064, India
| | - Jogin Desai
- Eyestem Research Private Limited, Centre for Cellular and Molecular Platforms (CCAMP), National Centre for Biological Sciences-Tata Institute of Fundamental Research (NCBS-TIFR), GKVK Campus, Bangalore, 560065, India; Centre for Eye Genetics and Research, Cytecare Hospital, Bellary Road, Bangalore, 560064, India
| | - Rajarshi Pal
- Eyestem Research Private Limited, Centre for Cellular and Molecular Platforms (CCAMP), National Centre for Biological Sciences-Tata Institute of Fundamental Research (NCBS-TIFR), GKVK Campus, Bangalore, 560065, India.
| |
Collapse
|