1
|
Barravecchia I, De Cesari C, Guadagni V, Signore G, Bertolini E, Giannelli SG, Scebba F, Martini D, Pè ME, Broccoli V, Andreazzoli M, Angeloni D, Demontis GC. Increasing cell culture density during a developmental window prevents fated rod precursors derailment toward hybrid rod-glia cells. Sci Rep 2023; 13:6025. [PMID: 37055439 PMCID: PMC10101963 DOI: 10.1038/s41598-023-32571-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 03/29/2023] [Indexed: 04/15/2023] Open
Abstract
In proliferating multipotent retinal progenitors, transcription factors dynamics set the fate of postmitotic daughter cells, but postmitotic cell fate plasticity driven by extrinsic factors remains controversial. Transcriptome analysis reveals the concurrent expression by postmitotic rod precursors of genes critical for the Müller glia cell fate, which are rarely generated from terminally-dividing progenitors as a pair with rod precursors. By combining gene expression and functional characterisation in single cultured rod precursors, we identified a time-restricted window where increasing cell culture density switches off the expression of genes critical for Müller glial cells. Intriguingly, rod precursors in low cell culture density maintain the expression of genes of rod and glial cell fate and develop a mixed rod/Muller glial cells electrophysiological fingerprint, revealing rods derailment toward a hybrid rod-glial phenotype. The notion of cell culture density as an extrinsic factor critical for preventing rod-fated cells diversion toward a hybrid cell state may explain the occurrence of hybrid rod/MG cells in the adult retina and provide a strategy to improve engraftment yield in regenerative approaches to retinal degenerative disease by stabilising the fate of grafted rod precursors.
Collapse
Affiliation(s)
- Ivana Barravecchia
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, 56126, Pisa, Italy
- Scuola Superiore Sant'Anna, Pisa, Italy
| | - Chiara De Cesari
- Scuola Superiore Sant'Anna, Pisa, Italy
- Department of Biology, University of Pisa, Pisa, Italy
| | | | - Giovanni Signore
- Department of Biology, University of Pisa, Pisa, Italy
- Fondazione Pisana per la Scienza, San Giuliano Terme, Italy
| | - Edoardo Bertolini
- Scuola Superiore Sant'Anna, Pisa, Italy
- Donald Danforth Plant Science Center, St. Louis, USA
| | | | | | | | | | - Vania Broccoli
- San Raffaele Hospital, Milan, Italy
- Institute of Neuroscience, National Research Council of Italy, Milan, Italy
| | | | | | - Gian Carlo Demontis
- Department of Pharmacy, University of Pisa, Via Bonanno Pisano, 6, 56126, Pisa, Italy.
| |
Collapse
|
2
|
Chen X, Li S, Liu X, Zhao J, Wu L, You R, Wang Y. Stimulation of C-Kit+ Retinal Progenitor Cells by Stem Cell Factor Confers Protection Against Retinal Degeneration. Front Pharmacol 2022; 13:796380. [PMID: 35431956 PMCID: PMC9008784 DOI: 10.3389/fphar.2022.796380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 02/28/2022] [Indexed: 11/13/2022] Open
Abstract
C-kit/CD117, expressed in a series of tissue-specific progenitor cells, plays an important role in tissue regeneration and tissue homeostasis. We previously demonstrated that organoid-derived c-kit+ retinal progenitor cells can facilitate the restoration of degenerated retina. Meanwhile, we have identified a population of endogenous c-kit+ cells in retinas of adult mouse. However, the exact role of these cells in retinal degeneration remains unclear. Here, we demonstrated that stimulation of endogenous c-kit+ cells by stem cell factor (SCF) conferred protection against retinal degeneration. Retinal degeneration was induced by intravitreal injection of N-methyl-D-aspartate (NMDA). NMDA challenge increased the total number of c-kit+ cells in the retinal ganglion cell layer (GCL), while deregulated the protein level of SCF, which was mainly expressed in Müller cells. Both flash electroretinogram (fERG) and light/dark transition tests showed that intravitreal injection of SCF effectively improved the visual function of NMDA-treated mice. Mechanistically, SCF administration not only prevented the loss of retinal ganglion cells (RGCs), but also maintained the function of RGCs as quantified by fERG. Further, we performed transcriptome sequencing analysis of the retinal cells isolated from SCF-treated mice and the parallel control. Gene Ontology analysis showed that SCF-induced transcriptome changes were closely correlated with eye development-related pathways. Crystallins and several protective factors such as Pitx3 were significantly upregulated by SCF treatment. Our results revealed the role of SCF stimulated c-kit+ cells in the protection of RGCs in NMDA-treated mice, via inhibiting the loss of RGCs. Administration of SCF can act as a potent strategy for treating retinal degeneration-related diseases.
Collapse
Affiliation(s)
- Xi Chen
- Department of Ophthalmology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- *Correspondence: Xi Chen, ; Yanling Wang,
| | - Shanshan Li
- Department of Ophthalmology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaoli Liu
- Department of Pediatric Newborn Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Jingjie Zhao
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lanting Wu
- Department of Ophthalmology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Ran You
- Department of Ophthalmology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yanling Wang
- Department of Ophthalmology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- *Correspondence: Xi Chen, ; Yanling Wang,
| |
Collapse
|
3
|
He XY, Zhao CJ, Xu H, Chen K, Bian BSJ, Gong Y, Weng CH, Zeng YX, Fu Y, Liu Y, Yin ZQ. Synaptic repair and vision restoration in advanced degenerating eyes by transplantation of retinal progenitor cells. Stem Cell Reports 2021; 16:1805-1817. [PMID: 34214489 PMCID: PMC8282465 DOI: 10.1016/j.stemcr.2021.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 12/25/2022] Open
Abstract
Stem cell transplantation shows enormous potential for treatment of incurable retinal degeneration (RD). To determine if and how grafts connect with the neural circuits of the advanced degenerative retina (ADR) and improve vision, we perform calcium imaging of GCaMP5-positive grafts in retinal slices. The organoid-derived C-Kit+/SSEA1- (C-Kit+) retinal progenitor cells (RPCs) become synaptically organized and build spontaneously active synaptic networks in three major layers of ADR. Light stimulation of the host photoreceptors elicits distinct neuronal responses throughout the graft RPCs. The graft RPCs and their differentiated offspring cells in inner nuclear layer synchronize their activities with the host cells and exhibit presynaptic calcium flux patterns that resemble intact retinal neurons. Once graft-to-host network is established, progressive vision loss is stabilized while control eyes continually lose vision. Therefore, transplantation of organoid-derived C-Kit+ RPCs can form functional synaptic networks within ADR and it holds promising avenue for advanced RD treatment.
Collapse
Affiliation(s)
- Xiang-Yu He
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Department of Ophthalmology, the 958th Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China; Department of Ophthalmology, General Hospital of Chinese People's Liberation Army, Beijing 100853, P.R. China
| | - Cong-Jian Zhao
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China
| | - Kang Chen
- Department of Ophthalmology, the 958th Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China
| | - Bai-Shi-Jiao Bian
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China
| | - Yu Gong
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China
| | - Chuan-Huang Weng
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China
| | - Yu-Xiao Zeng
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China
| | - Yan Fu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China
| | - Yong Liu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China.
| | - Zheng-Qin Yin
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, P.R. China; Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, P.R. China; Department of Ophthalmology, General Hospital of Chinese People's Liberation Army, Beijing 100853, P.R. China.
| |
Collapse
|
4
|
Li H, Lian L, Liu B, Chen Y, Yang J, Jian S, Zhou J, Xu Y, Ma X, Qu J, Hou L. KIT ligand protects against both light-induced and genetic photoreceptor degeneration. eLife 2020; 9:51698. [PMID: 32242818 PMCID: PMC7170656 DOI: 10.7554/elife.51698] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 04/02/2020] [Indexed: 12/25/2022] Open
Abstract
Photoreceptor degeneration is a major cause of blindness and a considerable health burden during aging but effective therapeutic or preventive strategies have not so far become readily available. Here, we show in mouse models that signaling through the tyrosine kinase receptor KIT protects photoreceptor cells against both light-induced and inherited retinal degeneration. Upon light damage, photoreceptor cells upregulate Kit ligand (KITL) and activate KIT signaling, which in turn induces nuclear accumulation of the transcription factor NRF2 and stimulates the expression of the antioxidant gene Hmox1. Conversely, a viable Kit mutation promotes light-induced photoreceptor damage, which is reversed by experimental expression of Hmox1. Furthermore, overexpression of KITL from a viral AAV8 vector prevents photoreceptor cell death and partially restores retinal function after light damage or in genetic models of human retinitis pigmentosa. Hence, application of KITL may provide a novel therapeutic avenue for prevention or treatment of retinal degenerative diseases.
Collapse
Affiliation(s)
- Huirong Li
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Lili Lian
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Bo Liu
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yu Chen
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Jinglei Yang
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Shuhui Jian
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiajia Zhou
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Ying Xu
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, China
| | - Xiaoyin Ma
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Jia Qu
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Ling Hou
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
5
|
Ahmad I, Teotia P, Erickson H, Xia X. Recapitulating developmental mechanisms for retinal regeneration. Prog Retin Eye Res 2019; 76:100824. [PMID: 31843569 DOI: 10.1016/j.preteyeres.2019.100824] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/06/2019] [Accepted: 12/11/2019] [Indexed: 12/18/2022]
Abstract
Degeneration of specific retinal neurons in diseases like glaucoma, age-related macular degeneration, and retinitis pigmentosa is the leading cause of irreversible blindness. Currently, there is no therapy to modify the disease-associated degenerative changes. With the advancement in our knowledge about the mechanisms that regulate the development of the vertebrate retina, the approach to treat blinding diseases through regenerative medicine appears a near possibility. Recapitulation of developmental mechanisms is critical for reproducibly generating cells in either 2D or 3D culture of pluripotent stem cells for retinal repair and disease modeling. It is the key for unlocking the neurogenic potential of Müller glia in the adult retina for therapeutic regeneration. Here, we examine the current status and potential of the regenerative medicine approach for the retina in the backdrop of developmental mechanisms.
Collapse
Affiliation(s)
- Iqbal Ahmad
- 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
| | - Helen Erickson
- Department of Ophthalmology and Visual Science, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Xiaohuan Xia
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200072, China
| |
Collapse
|
6
|
Ma X, Li H, Chen Y, Yang J, Chen H, Arnheiter H, Hou L. The transcription factor MITF in RPE function and dysfunction. Prog Retin Eye Res 2019; 73:100766. [DOI: 10.1016/j.preteyeres.2019.06.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/17/2019] [Accepted: 06/21/2019] [Indexed: 12/18/2022]
|
7
|
Gong Y, He X, Li Q, He J, Bian B, Li Y, Ge L, Zeng Y, Xu H, Yin ZQ. SCF/SCFR signaling plays an important role in the early morphogenesis and neurogenesis of human embryonic neural retina. Development 2019; 146:dev.174409. [PMID: 31548215 DOI: 10.1242/dev.174409] [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: 11/30/2018] [Accepted: 09/16/2019] [Indexed: 12/20/2022]
Abstract
The stem cell factor receptor (SCFR) has been demonstrated to be expressed in the neural retina of mice, rat and human for decades. Previous reports indicated that the SCFR correlates with glia differentiation of late retinal progenitor cells (RPCs), retinal vasculogenesis and homeostasis of the blood-retinal barrier. However, the role of SCF/SCFR signaling in the growth and development of the neural retina (NR), especially in the early embryonic stage, remains poorly understood. Here, we show that SCF/SCFR signaling orchestrates invagination of the human embryonic stem cell (hESC)-derived NR via regulation of cell cycle progression, cytoskeleton dynamic and apical constriction of RPCs in the ciliary marginal zone (CMZ). Furthermore, activation of SCF/SCFR signaling promotes neurogenesis in the central-most NR via acceleration of the migration of immature ganglion cells and repressing apoptosis. Our study reveals an unreported role for SCF/SCFR signaling in controlling ciliary marginal cellular behaviors during early morphogenesis and neurogenesis of the human embryonic NR, providing a new potential therapeutic target for human congenital eye diseases such as anophthalmia, microphthalmia and congenital high myopia.
Collapse
Affiliation(s)
- Yu Gong
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Xiangyu He
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Qiyou Li
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Juncai He
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Baishijiao Bian
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Yijian Li
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Linlin Ge
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Yuxiao Zeng
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Haiwei Xu
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China .,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| | - Zheng Qin Yin
- Southwest Hospital/ Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China .,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, PR China
| |
Collapse
|
8
|
Zou T, Gao L, Zeng Y, Li Q, Li Y, Chen S, Hu X, Chen X, Fu C, Xu H, Yin ZQ. Organoid-derived C-Kit +/SSEA4 - human retinal progenitor cells promote a protective retinal microenvironment during transplantation in rodents. Nat Commun 2019; 10:1205. [PMID: 30872578 PMCID: PMC6418223 DOI: 10.1038/s41467-019-08961-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 02/11/2019] [Indexed: 12/22/2022] Open
Abstract
Stem cell therapy may replace lost photoreceptors and preserve residual photoreceptors during retinal degeneration (RD). Unfortunately, the degenerative microenvironment compromises the fate of grafted cells, demanding supplementary strategies for microenvironment regulation. Donor cells with both proper regeneration capability and intrinsic ability to improve microenvironment are highly desired. Here, we use cell surface markers (C-Kit+/SSEA4−) to effectively eliminate tumorigenic embryonic cells and enrich retinal progenitor cells (RPCs) from human embryonic stem cell (hESC)-derived retinal organoids, which, following subretinal transplantation into RD models of rats and mice, significantly improve vision and preserve the retinal structure. We characterize the pattern of integration and materials transfer following transplantation, which likely contribute to the rescued photoreceptors. Moreover, C-Kit+/SSEA4− cells suppress microglial activation, gliosis and the production of inflammatory mediators, thereby providing a healthier host microenvironment for the grafted cells and delaying RD. Therefore, C-Kit+/SSEA4− cells from hESC-derived retinal organoids are a promising therapeutic cell source. Stem cell transplantation to treat retinal degeneration could be limited by the degenerative microenvironment. Here, the authors show that C-Kit+/SSEA4– progenitor cells enriched from human embryonic stem cell derived retinal organoids protect retinal structure, suppress microglial activation, gliosis and inflammation.
Collapse
Affiliation(s)
- Ting Zou
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Lixiong Gao
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Yuxiao Zeng
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Qiyou Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Yijian Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Siyu Chen
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Xisu Hu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Xi Chen
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Caiyun Fu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China. .,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China.
| | - Zheng Qin Yin
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China. .,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China.
| |
Collapse
|
9
|
Lin MS, Liao PY, Chen HM, Chang CP, Chen SK, Chern Y. Degeneration of ipRGCs in Mouse Models of Huntington's Disease Disrupts Non-Image-Forming Behaviors Before Motor Impairment. J Neurosci 2019; 39:1505-1524. [PMID: 30587542 PMCID: PMC6381252 DOI: 10.1523/jneurosci.0571-18.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 11/22/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022] Open
Abstract
Intrinsically photosensitive retinal ganglion cells (ipRGCs), which express the photopigment melanopsin, are photosensitive neurons in the retina and are essential for non-image-forming functions, circadian photoentrainment, and pupillary light reflexes. Five subtypes of ipRGCs (M1-M5) have been identified in mice. Although ipRGCs are spared in several forms of inherited blindness, they are affected in Alzheimer's disease and aging, which are associated with impaired circadian rhythms. Huntington's disease (HD) is an autosomal neurodegenerative disease caused by the expansion of a CAG repeat in the huntingtin gene. In addition to motor function impairment, HD mice also show impaired circadian rhythms and loss of ipRGC. Here, we found that, in HD mouse models (R6/2 and N171-82Q male mice), the expression of melanopsin was reduced before the onset of motor deficits. The expression of retinal T-box brain 2, a transcription factor essential for ipRGCs, was associated with the survival of ipRGCs. The number of M1 ipRGCs in R6/2 male mice was reduced due to apoptosis, whereas non-M1 ipRGCs were relatively resilient to HD progression. Most importantly, the reduced innervations of M1 ipRGCs, which was assessed by X-gal staining in R6/2-OPN4Lacz/+ male mice, contributed to the diminished light-induced c-fos and vasoactive intestinal peptide in the suprachiasmatic nuclei (SCN), which may explain the impaired circadian photoentrainment in HD mice. Collectively, our results show that M1 ipRGCs were susceptible to the toxicity caused by mutant Huntingtin. The resultant impairment of M1 ipRGCs contributed to the early degeneration of the ipRGC-SCN pathway and disrupted circadian regulation during HD progression.SIGNIFICANCE STATEMENT Circadian disruption is a common nonmotor symptom of Huntington's disease (HD). In addition to the molecular defects in the suprachiasmatic nuclei (SCN), the cause of circadian disruption in HD remains to be further explored. We hypothesized that ipRGCs, by integrating light input to the SCN, participate in the circadian regulation in HD mice. We report early reductions in melanopsin in two mouse models of HD, R6/2, and N171-82Q. Suppression of retinal T-box brain 2, a transcription factor essential for ipRGCs, by mutant Huntingtin might mediate the reduced number of ipRGCs. Importantly, M1 ipRGCs showed higher susceptibility than non-M1 ipRGCs in R6/2 mice. The resultant impairment of M1 ipRGCs contributed to the early degeneration of the ipRGC-SCN pathway and the circadian abnormality during HD progression.
Collapse
Affiliation(s)
- Meng-Syuan Lin
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei 115, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, and
| | - Po-Yu Liao
- Department of Life Science, National Taiwan University, Taipei 106, Taiwan
| | - Hui-Mei Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, and
| | - Ching-Pang Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, and
| | - Shih-Kuo Chen
- Department of Life Science, National Taiwan University, Taipei 106, Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115, Taiwan, and
| |
Collapse
|
10
|
Kuribayashi H, Baba Y, Iwagawa T, Arai E, Murakami A, Watanabe S. Roles of Nmnat1 in the survival of retinal progenitors through the regulation of pro-apoptotic gene expression via histone acetylation. Cell Death Dis 2018; 9:891. [PMID: 30166529 PMCID: PMC6117278 DOI: 10.1038/s41419-018-0907-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 07/16/2018] [Accepted: 07/19/2018] [Indexed: 11/21/2022]
Abstract
Leber congenital amaurosis (LCA) is a severe, genetically heterogeneous dystrophy of the retina and mutations in the nicotinamide mononucleotide adenylyltransferase 1 (NMNAT1) gene is one of causal factors of LCA. NMNAT1 is a nuclear enzyme essential for nicotinamide adenine dinucleotide (NAD) biosynthesis pathways, but the mechanisms underlying the LCA pathology and whether NMNAT1 has a role in normal retinal development remain unclear. Thus, we examined the roles of Nmnat1 in retinal development via short hairpin (sh)-RNA-mediated downregulation. Retinal explants expressing sh-Nmnat1 showed large numbers of apoptotic retinal progenitor cells in the inner half of the neuroblastic layer. Decreased intracellular NAD content was observed and the addition of NAD to the culture medium attenuated sh-Nmnat1-induced apoptosis. Of the nuclear Sirtuin (Sirt) family, the expression of sh-Sirt1 and sh-Sirt6 resulted in a phenotype similar to that of sh-Nmnat1. Sirt proteins are histone deacetylases and the expression of sh-Nmnat1 increased the levels of acetylated histones H3 and H4 in the retina. Expression of sh-Nmnat1 resulted in significantly increased expression of Noxa and Fas, two pro-apoptotic genes. Acetylation of the genomic 5′-untranslated regions of Noxa and Fas loci was upregulated by sh-Nmnat1 expression. The co-expression of sh-Fas with sh-Nmnat1 reduced the number of apoptotic cells induced by sh-Nmnat1 expression alone. Taken together, our data suggested that the increased expression of Noxa and Fas explains, at least in part, the phenotype associated with sh-Nmnat1 in the retina. Taken together, these findings demonstrate the importance of the NAD biosynthesis pathway in normal development of the retina.
Collapse
Affiliation(s)
- Hiroshi Kuribayashi
- Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yukihiro Baba
- Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Toshiro Iwagawa
- Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Eisuke Arai
- Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan.,Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Akira Murakami
- Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Sumiko Watanabe
- Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
| |
Collapse
|
11
|
Suzuki-Kerr H, Iwagawa T, Sagara H, Mizota A, Suzuki Y, Watanabe S. Pivotal roles of Fezf2 in differentiation of cone OFF bipolar cells and functional maturation of cone ON bipolar cells in retina. Exp Eye Res 2018; 171:142-154. [DOI: 10.1016/j.exer.2018.03.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Revised: 03/05/2018] [Accepted: 03/16/2018] [Indexed: 10/17/2022]
|
12
|
Chen X, Wang S, Xu H, Pereira JD, Hatzistergos KE, Saur D, Seidler B, Hare JM, Perrella MA, Yin ZQ, Liu X. Evidence for a retinal progenitor cell in the postnatal and adult mouse. Stem Cell Res 2017; 23:20-32. [PMID: 28672156 DOI: 10.1016/j.scr.2017.06.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 06/16/2017] [Accepted: 06/19/2017] [Indexed: 12/22/2022] Open
Abstract
Progress in cell therapy for retinal disorders has been challenging. Recognized retinal progenitors are a heterogeneous population of cells that lack surface markers for the isolation of live cells for clinical implementation. In the present application, our objective was to use the stem cell factor receptor c-Kit (CD117), a surface marker, to isolate and evaluate a distinct progenitor cell population from retinas of postnatal and adult mice. Here we report that, by combining traditional methods with fate mapping, we have identified a c-Kit-positive (c-Kit+) retinal progenitor cell (RPC) that is self-renewing and clonogenic in vitro, and capable of generating many cell types in vitro and in vivo. Based on cell lineage tracing, significant subpopulations of photoreceptors in the outer nuclear layer and bipolar, horizontal, amacrine and Müller cells in the inner nuclear layer are the progeny of c-Kit+ cells in vivo. The RPC progeny contributes to retinal neurons and glial cells, which are responsible for the conversion of light into visual signals. The ability to isolate and expand in vitro live c-Kit+ RPCs makes them a future therapeutic option for retinal diseases.
Collapse
Affiliation(s)
- Xi Chen
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, China; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shaojun Wang
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, China; Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, China
| | - Joao D Pereira
- Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Anesthesia, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Dieter Saur
- Medicine II, Technische Universitaet Muenchen, Munich, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Barbara Seidler
- Medicine II, Technische Universitaet Muenchen, Munich, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Joshua M Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mark A Perrella
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zheng Qin Yin
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, China
| | - Xiaoli Liu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pediatric Newborn Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Chen X, Chen Z, Li Z, Zhao C, Zeng Y, Zou T, Fu C, Liu X, Xu H, Yin ZQ. Grafted c-kit +/SSEA1 - eye-wall progenitor cells delay retinal degeneration in mice by regulating neural plasticity and forming new graft-to-host synapses. Stem Cell Res Ther 2016; 7:191. [PMID: 28038685 PMCID: PMC5203726 DOI: 10.1186/s13287-016-0451-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 11/25/2016] [Accepted: 12/06/2016] [Indexed: 12/12/2022] Open
Abstract
Background Despite diverse pathogenesis, the common pathological change observed in age-related macular degeneration and in most hereditary retinal degeneration (RD) diseases is photoreceptor loss. Photoreceptor replacement by cell transplantation may be a feasible treatment for RD. The major obstacles to clinical translation of stem cell-based cell therapy in RD remain the difficulty of obtaining sufficient quantities of appropriate and safe donor cells and the poor integration of grafted stem cell-derived photoreceptors into the remaining retinal circuitry. Methods Eye-wall c-kit+/stage-specific embryonic antigen 1 (SSEA1)− cells were isolated via fluorescence-activated cell sorting, and their self-renewal and differentiation potential were detected by immunochemistry and flow cytometry in vitro. After labeling with quantum nanocrystal dots and transplantation into the subretinal space of rd1 RD mice, differentiation and synapse formation by daughter cells of the eye-wall c-kit+/SSEA1− cells were evaluated by immunochemistry and western blotting. Morphological changes of the inner retina of rd1 mice after cell transplantation were demonstrated by immunochemistry. Retinal function of rd1 mice that received cell grafts was tested via flash electroretinograms and the light/dark transition test. Results Eye-wall c-kit+/SSEA1− cells were self-renewing and clonogenic, and they retained their proliferative potential through more than 20 passages. Additionally, eye-wall c-kit+/SSEA1− cells were capable of differentiating into multiple retinal cell types including photoreceptors, bipolar cells, horizontal cells, amacrine cells, Müller cells, and retinal pigment epithelium cells and of transdifferentiating into smooth muscle cells and endothelial cells in vitro. The levels of synaptophysin and postsynaptic density-95 in the retinas of eye-wall c-kit+/SSEA1− cell-transplanted rd1 mice were significantly increased at 4 weeks post transplantation. The c-kit+/SSEA1− cells were capable of differentiating into functional photoreceptors that formed new synaptic connections with recipient retinas in rd1 mice. Transplantation also partially corrected the abnormalities of inner retina of rd1 mice. At 4 and 8 weeks post transplantation, the rd1 mice that received c-kit+/SSEA1− cells showed significant increases in a-wave and b-wave amplitude and the percentage of time spent in the dark area. Conclusions Grafted c-kit+/SSEA1− cells restored the retinal function of rd1 mice via regulating neural plasticity and forming new graft-to-host synapses. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0451-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xi Chen
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China.,School of Medicine, Nankai University, Tianjin, 300071, China.,Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Zehua Chen
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Zhengya Li
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Chen Zhao
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Yuxiao Zeng
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Ting Zou
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Caiyun Fu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China
| | - Xiaoli Liu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.,Department of Pediatric Newborn Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China. .,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China.
| | - Zheng Qin Yin
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China. .,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing, 400038, China.
| |
Collapse
|
14
|
Koso H, Tsuhako A, Lai CY, Baba Y, Otsu M, Ueno K, Nagasaki M, Suzuki Y, Watanabe S. Conditional rod photoreceptor ablation reveals Sall1 as a microglial marker and regulator of microglial morphology in the retina. Glia 2016; 64:2005-24. [PMID: 27459098 DOI: 10.1002/glia.23038] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 06/10/2016] [Accepted: 07/07/2016] [Indexed: 12/18/2022]
Abstract
Neurodegeneration has been shown to induce microglial activation and the infiltration of monocyte-derived macrophages into the CNS, resulting in the coexistence of these two populations within the same lesion, though their distinct features remain elusive. To investigate the impact of rod photoreceptor degeneration on microglial activation, we generated a toxin-mediated genetic model of rod degeneration. Rod injury induced microglial proliferation and migration toward the photoreceptors. Bone marrow transplantation revealed the invasion of monocyte-derived macrophages into the retina, with microglia and the infiltrating macrophages showing distinct distribution patterns in the retina. By comparing the gene expression profiles of the activated microglia and infiltrating macrophages, we identified microglia-specific genes, including Ak1, Ctsf, Sall1, Phlda3, and Spns2. An analysis of Sall1gfp knock-in mice showed GFP expression in the microglia of developing and mature healthy retinas. DTA injury induced the expansion of Sall1gfp(+) microglia, whereas Ly6C(+) monocyte-derived macrophages were mostly Sall1gfp(-) , supporting the idea that Sall1 is exclusively expressed in microglia within the retinal phagocyte pool. We evaluated the contribution of microglia to the phagocyte pool in rd1 mutant retinas and found that Sall1gfp(+) microglia constituted the majority of phagocytes. A Sall1 deficiency did not affect microglial colonization of the retina and the cortex, but it did change their morphology from a ramified to a more amoeboid appearance. The morphological defects observed in Sall1-deficient microglia were not rescued by the presence of wild-type non-microglial cells, suggesting that Sall1 functions cell-autonomously in microglia. Taken together, our data indicate that Sall1 regulates microglial morphology during development. GLIA 2016;64:2005-2024.
Collapse
Affiliation(s)
- Hideto Koso
- Division of Molecular and Developmental Biology, Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Asano Tsuhako
- Division of Molecular and Developmental Biology, Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Chen-Yi Lai
- Division of Stem Cell Processing and Stem Cell Bank, Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Yukihiro Baba
- Division of Molecular and Developmental Biology, Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Makoto Otsu
- Division of Stem Cell Processing and Stem Cell Bank, Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Kazuko Ueno
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Masao Nagasaki
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Yutaka Suzuki
- Department of Bioinformatics and Systems Biology, Graduate School of Frontier Sciences, the University of Tokyo, Kashiwa, Japan
| | - Sumiko Watanabe
- Division of Molecular and Developmental Biology, Institute of Medical Science, the University of Tokyo, Tokyo, Japan.
| |
Collapse
|
15
|
Zhou PY, Peng GH, Xu H, Yin ZQ. c-Kit+ cells isolated from human fetal retinas represent a new population of retinal progenitor cells. J Cell Sci 2015; 128:2169-78. [PMID: 25918122 DOI: 10.1242/jcs.169086] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/20/2015] [Indexed: 12/26/2022] Open
Abstract
ABSTRACT
Definitive surface markers for retinal progenitor cells (RPCs) are still lacking. Therefore, we sorted c-Kit+ and stage-specific embryonic antigen-4− (SSEA4−) retinal cells for further biological characterization. RPCs were isolated from human fetal retinas (gestational age of 12–14 weeks). c-Kit+/SSEA4− RPCs were sorted by fluorescence-activated cell sorting, and their proliferation and differentiation capabilities were evaluated by using immunocytochemistry and flow cytometry. The effectiveness and safety were assessed following injection of c-Kit+/SSEA4− cells into the subretina of Royal College of Surgeons (RCS) rats. c-Kit+ cells were found in the inner part of the fetal retina. Sorted c-Kit+/SSEA4− cells expressed retinal stem cell markers. Our results clearly demonstrate the proliferative potential of these cells. Moreover, c-Kit+/SSEA4− cells differentiated into retinal cells that expressed markers of photoreceptor cells, ganglion cells and glial cells. These cells survived for at least 3 months after transplantation into the host subretinal space. Teratomas were not observed in the c-Kit+/SSEA4−-cell group. Thus, c-Kit can be used as a surface marker for RPCs, and c-Kit+/SSEA4− RPCs exhibit the ability to self-renew and differentiate into retinal cells.
Collapse
Affiliation(s)
- Peng-Yi Zhou
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, He'nan 450003, China
| | - Guang-Hua Peng
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, He'nan 450003, China
- Department of Ophthalmology, General Hospital of Chinese People's Liberation Army, Beijing 100853, China
| | - Haiwei Xu
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China
- Key Lab of Ophthalmology of Chinese People's Liberation Army, Chongqing 400038, China
| | - Zheng Qin Yin
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China
- Key Lab of Ophthalmology of Chinese People's Liberation Army, Chongqing 400038, China
| |
Collapse
|
16
|
Enright JM, Lawrence KA, Hadzic T, Corbo JC. Transcriptome profiling of developing photoreceptor subtypes reveals candidate genes involved in avian photoreceptor diversification. J Comp Neurol 2014; 523:649-68. [PMID: 25349106 DOI: 10.1002/cne.23702] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 10/21/2014] [Accepted: 10/22/2014] [Indexed: 12/26/2022]
Abstract
Avian photoreceptors are a diverse class of neurons, comprised of four single cones, the two members of the double cone, and rods. The signaling events and transcriptional regulators driving the differentiation of these diverse photoreceptors are largely unknown. In addition, many distinctive features of photoreceptor subtypes, including spectral tuning, oil droplet size and pigmentation, synaptic targets, and spatial patterning, have been well characterized, but the molecular mechanisms underlying these attributes have not been explored. To identify genes specifically expressed in distinct chicken (Gallus gallus) photoreceptor subtypes, we developed fluorescent reporters that label photoreceptor subpopulations, isolated these subpopulations by using fluorescence-activated cell sorting, and subjected them to next-generation sequencing. By comparing the expression profiles of photoreceptors labeled with rhodopsin, red opsin, green opsin, and violet opsin reporters, we have identified hundreds of differentially expressed genes that may underlie the distinctive features of these photoreceptor subtypes. These genes are involved in a variety of processes, including phototransduction, transcriptional regulation, cell adhesion, maintenance of intra- and extracellular structure, and metabolism. Of particular note are a variety of differentially expressed transcription factors, which may drive and maintain photoreceptor diversity, and cell adhesion molecules, which may mediate spatial patterning of photoreceptors and act to establish retinal circuitry. These analyses provide a framework for future studies that will dissect the role of these various factors in the differentiation of avian photoreceptor subtypes.
Collapse
Affiliation(s)
- Jennifer M Enright
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, 63110-1024
| | | | | | | |
Collapse
|
17
|
Kuribayashi H, Baba Y, Watanabe S. BMP signaling participates in late phase differentiation of the retina, partly via upregulation of Hey2. Dev Neurobiol 2014; 74:1172-83. [PMID: 24890415 DOI: 10.1002/dneu.22196] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 04/25/2014] [Accepted: 05/24/2014] [Indexed: 11/11/2022]
Abstract
Bone morphogenetic protein (BMP) plays pivotal roles in early retinal development. However, its roles in the late phase of retinal development remain unclear. We found that BMP receptors and ligands were expressed in the postnatal mouse retina. Furthermore, immunostaining revealed that phosphorylated Smads were enriched in various cells types in the inner nuclear layer postnatally. However, phosphorylated Smads were not detected in photoreceptors, suggesting that BMP may play roles in retinal cells in the inner nuclear layer. Forced expression of constitutively active BMP receptors during retinal development resulted in an increased number of bipolar cells and Müller glia and a decreased number of rod photoreceptors; however, proliferation was not perturbed. The expression of dominant negative BMP receptors resulted in a decreased number of Müller glia and bipolar cells. In addition, inhibiting BMP signaling in retinal monolayer cultures abrogated Müller glial process extension, suggesting that BMP signaling also plays a role in the maturation of Müller glia. The expression of the basic helix-loop-helix transcription factor Hey2 was induced by BMP signaling in retinas. The coexpression of sh-Hey2 with constitutively active BMP receptors suggested that the effects of BMP signaling on retinal differentiation could be attributed partly to the induction of Hey2 by BMP. We propose that BMP signaling plays pivotal roles in the differentiation of retinal progenitor cells into late differentiating retinal cell types and in the maturation of Müller glia; these effects were mediated, at least in part, by Hey2.
Collapse
Affiliation(s)
- Hiroshi Kuribayashi
- Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | | |
Collapse
|
18
|
Suga A, Sadamoto K, Fujii M, Mandai M, Takahashi M. Proliferation potential of Müller glia after retinal damage varies between mouse strains. PLoS One 2014; 9:e94556. [PMID: 24747725 PMCID: PMC3991641 DOI: 10.1371/journal.pone.0094556] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 03/18/2014] [Indexed: 01/12/2023] Open
Abstract
Retinal Müller glia can serve as a source for regeneration of damaged retinal neurons in fish, birds and mammals. However, the proliferation rate of Müller glia has been reported to be low in the mammalian retina. To overcome this problem, growth factors and morphogens have been studied as potent promoters of Müller glial proliferation, but the molecular mechanisms that limit the proliferation of Müller glia in the mammalian retina remain unknown. In the present study, we found that the degree of damage-induced Müller glia proliferation varies across mouse strains. In mouse line 129×1/SvJ (129), there was a significantly larger proliferative response compared with that observed in C57BL/6 (B6) after photoreceptor cell death. Treatment with a Glycogen synthase kinase 3 (GSK3) inhibitor enhanced the proliferation of Müller glia in 129 but not in B6 mouse retinas. We therefore focused on the different gene expression patterns during retinal degeneration between B6 and 129. Expression levels of Cyclin D1 and Nestin correlated with the degree of Müller glial proliferation. A comparison of genome-wide gene expression between B6 and 129 showed that distinct sets of genes were upregulated in the retinas after damage, including immune response genes and chromatin remodeling factors.
Collapse
Affiliation(s)
- Akiko Suga
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, Minatojima, Chu-O-ku, Kobe, Japan
| | - Kazuyo Sadamoto
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, Minatojima, Chu-O-ku, Kobe, Japan
| | - Momo Fujii
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, Minatojima, Chu-O-ku, Kobe, Japan
| | - Michiko Mandai
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, Minatojima, Chu-O-ku, Kobe, Japan
| | - Masayo Takahashi
- Laboratory for Retinal Regeneration, Center for Developmental Biology, RIKEN, Minatojima, Chu-O-ku, Kobe, Japan
- * E-mail:
| |
Collapse
|
19
|
Steven Shaw SW. Amniotic fluid stem cells for minimally invasive prenatal cell therapy. Gynecol Minim Invasive Ther 2014. [DOI: 10.1016/j.gmit.2014.01.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
20
|
Mochizuki Y, Iida A, Lyons E, Kageyama R, Nakauchi H, Murakami A, Watanabe S. Use of cell type-specific transcriptome to identify genes specifically involved in Müller glia differentiation during retinal development. Dev Neurobiol 2013; 74:426-37. [PMID: 24124169 DOI: 10.1002/dneu.22131] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Revised: 08/07/2013] [Accepted: 09/12/2013] [Indexed: 11/11/2022]
Abstract
Retinal progenitor cells alter their properties over the course of development, and sequentially produce different sub-populations of retinal cells. We had previously found that early and late retinal progenitor cell populations can be distinguished by their surface antigens, SSEA-1 and c-kit, respectively. Using DNA microarray analysis, we examined the transcriptomes of SSEA-1 positive cells at E14, and c-kit positive, and c-kit negative cells at P1. By comparing data, we identified genes specifically expressed in c-kit positive late retinal progenitor cells. The previous literature suggests that most of the c-kit positive cell-specific genes are related to glia differentiation in brain or are expressed in Müller glia. Since Notch signaling promotes Müller glia differentiation in retina, we examined the effects of gain- and loss-of-Notch signaling on expression of these genes and found that all the genes were positively affected by Notch signaling. Finally, we screened the genes for their function in retinal development by shRNA-based suppression in retinal explants. In about half the genes, Müller glia differentiation was perturbed when their expression was suppressed. Taken together, these results show that at P1, c-kit positive retinal progenitor cells, which include Müller glia precursor cells, are enriched for genes related to glial differentiation. We propose analysis of purified subsets of retinal cells as a powerful tool to elucidate the molecular basis of retinal development.
Collapse
Affiliation(s)
- Yujin Mochizuki
- Department of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan; Department of Ophthalmology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
21
|
Veréb Z, Lumi X, Andjelic S, Globocnik-Petrovic M, Urbancic M, Hawlina M, Facskó A, Petrovski G. Functional and molecular characterization of ex vivo cultured epiretinal membrane cells from human proliferative diabetic retinopathy. BIOMED RESEARCH INTERNATIONAL 2013; 2013:492376. [PMID: 24195074 PMCID: PMC3806336 DOI: 10.1155/2013/492376] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 08/13/2013] [Accepted: 08/15/2013] [Indexed: 12/13/2022]
Abstract
Characterization of the cell surface marker phenotype of ex vivo cultured cells growing out of human fibrovascular epiretinal membranes (fvERMs) from proliferative diabetic retinopathy (PDR) can give insight into their function in immunity, angiogenesis, and retinal detachment. FvERMs from uneventful vitrectomies due to PDR were cultured adherently ex vivo. Surface marker analysis, release of immunity- and angiogenesis-pathway-related factors upon TNF α activation and measurement of the intracellular calcium dynamics upon mechano-stimulation using fluorescent dye Fura-2 were all performed. FvERMs formed proliferating cell monolayers when cultured ex vivo, which were negative for endothelial cell markers (CD31, VEGFR2), partially positive for hematopoietic- (CD34, CD47) and mesenchymal stem cell markers (CD73, CD90/Thy-1, and PDGFR β ), and negative for CD105. CD146/MCAM and CD166/ALCAM, previously unreported in cells from fvERMs, were also expressed. Secretion of 11 angiogenesis-related factors (DPPIV/CD26, EG-VEGF/PK1, ET-1, IGFBP-2 and 3, IL-8/CXCL8, MCP-1/CCL2, MMP-9, PTX3/TSG-14, Serpin E1/PAI-1, Serpin F1/PEDF, TIMP-1, and TSP-1) were detected upon TNF α activation of fvERM cells. Mechano-stimulation of these cells induced intracellular calcium propagation representing functional viability and role of these cells in tractional retinal detachment, thus serving as a model for studying tractional forces present in fvERMs in PDR ex vivo.
Collapse
Affiliation(s)
- Zoltán Veréb
- Stem Cells and Eye Research Laboratory, Department of Biochemistry and Molecular Biology, Medical and Health Science Center, Faculty of Medicine, University of Debrecen, Debrecen H-4010, Hungary
| | - Xhevat Lumi
- Eye Hospital, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Sofija Andjelic
- Eye Hospital, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia
| | | | - Mojca Urbancic
- Eye Hospital, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Marko Hawlina
- Eye Hospital, University Medical Centre Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Andrea Facskó
- Department of Ophthalmology, University of Szeged, H-6720, Hungary
| | - Goran Petrovski
- Stem Cells and Eye Research Laboratory, Department of Biochemistry and Molecular Biology, Medical and Health Science Center, Faculty of Medicine, University of Debrecen, Debrecen H-4010, Hungary
- Department of Ophthalmology, University of Szeged, H-6720, Hungary
| |
Collapse
|
22
|
Usui A, Mochizuki Y, Iida A, Miyauchi E, Satoh S, Sock E, Nakauchi H, Aburatani H, Murakami A, Wegner M, Watanabe S. The early retinal progenitor-expressed gene Sox11 regulates the timing of the differentiation of retinal cells. Development 2013; 140:740-50. [PMID: 23318640 DOI: 10.1242/dev.090274] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Sry-related HMG box (Sox) proteins, Sox11 and Sox4 are members of the SoxC subtype. We found that Sox11 was strongly expressed in early retinal progenitor cells and that Sox4 expression began around birth, when expression of Sox11 subsided. To analyze the roles of Sox11 and Sox4 in retinal development, we perturbed their expression patterns in retinal explant cultures. Overexpression of Sox11 and Sox4 in retinal progenitors resulted in similar phenotypes: an increased number of cone cells and dramatically decreased numbers of rod cells and Müller glia. Birth-date analysis showed that cone cells were produced at a later developmental stage than that in which cone genesis normally occurs. Sox11-knockout retinas showed delayed onset and progress of differentiation of subsets of retinal cells during the embryonic period. After birth, retinal differentiation took place relatively normally, probably because of the redundant activity of Sox4, which starts to be expressed around birth. Overexpression and loss-of-function analysis failed to provide any evidence that Sox11 and Sox4 directly regulate the transcription of genes crucial to the differentiation of subsets of retinal cells. However, histone H3 acetylation of some early proneural genes was reduced in knockout retina. Thus, Sox11 may create an epigenetic state that helps to establish the competency to differentiate. Taking our findings together, we propose that the sequential expression of Sox11 and Sox4 during retinogenesis leads to the fine adjustment of retinal differentiation by helping to establish the competency of retinal progenitors.
Collapse
Affiliation(s)
- Ayumi Usui
- Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Usui A, Iwagawa T, Mochizuki Y, Iida A, Wegner M, Murakami A, Watanabe S. Expression of Sox4 and Sox11 is regulated by multiple mechanisms during retinal development. FEBS Lett 2013; 587:358-63. [PMID: 23313252 DOI: 10.1016/j.febslet.2012.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 12/13/2012] [Accepted: 12/14/2012] [Indexed: 11/19/2022]
Abstract
Sox11 and Sox4 play critical roles in retinal development, during which they display specific and unique expression patterns. The expression of Sox11 and Sox4 is temporally sequential, albeit spatially overlapping in some retinal subtypes. Gain-of-function and loss-of-function analyses suggested that Notch signaling suppresses Sox4 expression in the early developing retina but not during the later period of development. The levels of histone H3-acetylation and H3-lysine 4 tri-methylation at the Sox11 locus declined during development, as did the levels of Sox11. A similar but less marked change was seen for Sox4. For both genes, histone H3-lysine 27 methylation was low during development and increased markedly in the adult.
Collapse
Affiliation(s)
- Ayumi Usui
- Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
24
|
Salm S, Burger PE, Wilson EL. TGF-β and stem cell factor regulate cell proliferation in the proximal stem cell niche. Prostate 2012; 72:998-1005. [PMID: 22024978 PMCID: PMC3275683 DOI: 10.1002/pros.21505] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 09/29/2011] [Indexed: 11/07/2022]
Abstract
BACKGROUND Stem cells are located in specific regulatory environments termed niches, which modulate the survival and proliferation of the cells through a variety of both mitogenic and inhibitory cytokines. In the murine prostate, stem cells are located in the proximal region of prostatic ducts. We examined the regulation of murine prostate cells in the stem cell niche by transforming growth factor beta (TGF-β) and stem cell factor (SCF). METHODS Prostate cells from the proximal and distal regions of prostatic ducts were cultured in the presence and absence of TGF-β and SCF, both on collagen-coated wells and in collagen gels. Cell growth on collagen was assessed by determining cell number. Cell growth in collagen gels was quantified by determining the number, size and complexity of prostatic ducts. The basal and luminal phenotype of the cells was determined by immunohistochemistry. RESULTS Endogenous TGF-β inhibited proliferation and promoted differentiation of proximal cells towards a luminal phenotype. It also inhibited duct-forming capacity and promoted differentiation of prostatic ducts towards a luminal phenotype. Addition of SCF enhanced proximal cell proliferation on collagen-coated wells and duct formation in collagen gels. Proliferation was further increased by ablation of endogenous TGF-β. CONCLUSION Proliferation and the basal/luminal cell composition of cells isolated from the proximal region of prostatic ducts, the stem cell niche, is regulated in part by opposing effects of SCF and endogenous TGF-β.
Collapse
Affiliation(s)
- Sarah Salm
- Department of Cell Biology, New York University School of Medicine, New York, New York
- Borough of Manhattan Community College, New York, New York
| | - Patricia E. Burger
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - E. Lynette Wilson
- Department of Cell Biology, New York University School of Medicine, New York, New York
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- New York University Cancer Institute, New York University School of Medicine, New York, New York
- Department of Urology, New York University School of Medicine, New York, New York
- Correspondence to E. Lynette Wilson, New York University School of Medicine, 550 First Avenue, NY, NY 10016, USA, , tel: 212-263-7684
| |
Collapse
|
25
|
Characterization of Progenitor Cells during Canine Retinal Development. Stem Cells Int 2012; 2012:675805. [PMID: 22567026 PMCID: PMC3328336 DOI: 10.1155/2012/675805] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Revised: 11/28/2011] [Accepted: 11/28/2011] [Indexed: 01/16/2023] Open
Abstract
We identify the presence of progenitor cells during retinal development in the dog, as this species represents a natural model for studying several breed-specific degenerative retinal disorders. Antibodies to detected progenitor cells (Pax6, C-kit, and nestin) and ganglion cells (BDNF, Brn3a, and Thy1) were used in combination with H3 for the purpose of identifying proliferating cells. Pax6, nestin, C-kit, and H3 were localized mainly in the neuroblastic layer of the retina during the embryonic stage. During the fetal stage, proteins were expressed in the inner neuroblastic layer (INL) as well as in the outer neuroblastic layer; BDNF, Thy1, and Brn3a were also expressed in the INL. During the neonatal stage only C-kit was not expressed. Proliferating cells were present in both undifferentiated and differentiated retina. These results suggest that, during canine retinogenesis, progenitor cells are distributed along the retina and some of these cells remain as progenitor cells of the ganglion cells during the first postnatal days.
Collapse
|
26
|
Iacono E, Brunori L, Pirrone A, Pagliaro PP, Ricci F, Tazzari PL, Merlo B. Isolation, characterization and differentiation of mesenchymal stem cells from amniotic fluid, umbilical cord blood and Wharton's jelly in the horse. Reproduction 2012; 143:455-68. [PMID: 22274885 DOI: 10.1530/rep-10-0408] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Mesenchymal stem cells (MSCs) have been derived from multiple sources of the horse including umbilical cord blood (UCB) and amnion. This work aimed to identify and characterize stem cells from equine amniotic fluid (AF), CB and Wharton's Jelly (WJ). Samples were obtained from 13 mares at labour. AF and CB cells were isolated by centrifugation, while WJ was prepared by incubating with an enzymatic solution for 2 h. All cell lines were cultured in DMEM/TCM199 plus fetal bovine serum. Fibroblast-like cells were observed in 7/10 (70%) AF, 6/8 (75%) CB and 8/12 (66.7%) WJ samples. Statistically significant differences were found between cell-doubling times (DTs): cells isolated from WJ expanded more rapidly (2.0±0.6 days) than those isolated from CB (2.6±1.3 days) and AF (2.3±1.0 days) (P<0.05). Positive von Kossa and Alizarin Red S staining confirmed osteogenesis. Alcian Blue staining of matrix glycosaminoglycans illustrated chondrogenesis and positive Oil Red O lipid droplets staining suggested adipogenesis. All cell lines isolated were positive for CD90, CD44, CD105; and negative for CD34, CD14 and CD45. These findings suggest that equine MSCs from AF, UCB and WJ appeared to be a readily obtainable and highly proliferative cell lines from a uninvasive source that may represent a good model system for stem cell biology and cellular therapy applications in horses. However, to assess their use as an allogenic cell source, further studies are needed for evaluating the expression of markers related to cell immunogenicity.
Collapse
Affiliation(s)
- Eleonora Iacono
- Department of Veterinary Medical Sciences, University of Bologna, via Tolara di Sopra 50, 40064 Ozzano Emilia, Bologna, Italy.
| | | | | | | | | | | | | |
Collapse
|
27
|
Saito R, Nakauchi H, Watanabe S. Serine/threonine kinase, Melk, regulates proliferation and glial differentiation of retinal progenitor cells. Cancer Sci 2012; 103:42-9. [PMID: 21923749 PMCID: PMC11164171 DOI: 10.1111/j.1349-7006.2011.02104.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Serine/threonine kinase, Melk, was initially cloned in oocytes, but it is expressed in normal tissues and especially in cancer cells. We had previously identified Melk as a gene that is highly expressed in immature mouse retinal progenitors. To analyze the function of Melk in embryogenesis, we cloned zebrafish Melk and reported that morpholino-based downregulation of Melk in zebrafish resulted in severe anemia. Melk-morpholino-treated zebrafish also showed microphthalmia, suggesting the participation of Melk in retinal development. In Melk-depleted retinas, differentiation of retinal neurons took place but was delayed, and the proliferative period of retinal progenitor cells was prolonged, suggesting that Melk might regulate the timing of the transition from proliferation to differentiation. For more detailed examination, we performed gain- and loss-of-function analyses of Melk in mouse retinas. Knockdown of Melk by shRNA in mouse embryonic retinal explant culture resulted in decreased proliferative activity of retinal progenitors, and accordingly, overexpression of Melk slightly enhanced proliferation. Differentiation of retinal progenitor into subtypes of retinal neurons was not significantly affected, but Müller glia differentiation was perturbed by the level of Melk. Furthermore, process extension of glial cells was enhanced in the absence of Melk, suggesting that Melk is involved in the morphological differentiation of retinal cells. Taken together, our results suggest that Melk is primarily required for proper proliferation, and might play multiple roles in retinal development in vertebrates.
Collapse
Affiliation(s)
- Rika Saito
- Department of Molecular and Developmental Biology, University of Tokyo, Tokyo, Japan
| | | | | |
Collapse
|
28
|
The role of Zic family zinc finger transcription factors in the proliferation and differentiation of retinal progenitor cells. Biochem Biophys Res Commun 2011; 415:42-7. [PMID: 22024047 DOI: 10.1016/j.bbrc.2011.10.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 10/03/2011] [Indexed: 02/07/2023]
Abstract
Members of the Zic family of zinc finger transcription factors play critical roles in a variety of developmental processes. Using DNA microarray analysis, we found that Zics are strongly expressed in SSEA-1-positive early retinal progenitors in the peripheral region of the mouse retina. Reverse-transcription polymerase chain reaction using mRNA from the retina at various developmental stages showed that Zic1 and Zic2 are expressed in the embryonic retina and then gradually disappear during retinal development. Zic3 is also expressed in the embryonic retina; its expression level slightly decreases but it is expressed until adulthood. We overexpressed Zic1, Zic2, or Zic3 in retinal progenitors at embryonic day 17.5 and cultured the retina as explants for 2 weeks. The number of rod photoreceptors was fewer than in the control, but no other cell types showed significant differences between control and Zic overexpressing cells. The proliferation activity of normal retinal progenitors decreased after 5 days in culture, as observed in normal in vivo developmental processes. However, Zic expressing retinal cells continued to proliferate at days 5 and 7, suggesting that Zics sustain the proliferation activities of retinal progenitor cells. Since the effects of Zic1, 2, and 3 are indistinguishable in terms of differentiation and proliferation of retinal progenitors, the redundant function of Zics in retinal development is suggested.
Collapse
|
29
|
Lakowski J, Han YT, Pearson RA, Gonzalez-Cordero A, West EL, Gualdoni S, Barber AC, Hubank M, Ali RR, Sowden JC. Effective transplantation of photoreceptor precursor cells selected via cell surface antigen expression. Stem Cells 2011; 29:1391-404. [PMID: 21774040 PMCID: PMC3303132 DOI: 10.1002/stem.694] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Retinal degenerative diseases are a major cause of untreatable blindness. Stem cell therapy to replace lost photoreceptors represents a feasible future treatment. We previously demonstrated that postmitotic photoreceptor precursors expressing an NrlGFP transgene integrate into the diseased retina and restore some light sensitivity. As genetic modification of precursor cells derived from stem cell cultures is not desirable for therapy, we have tested cell selection strategies using fluorochrome-conjugated antibodies recognizing cell surface antigens to sort photoreceptor precursors. Microarray analysis of postnatal NrlGFP-expressing precursors identified four candidate genes encoding cell surface antigens (Nt5e, Prom1, Podxl, and Cd24a). To test the feasibility of using donor cells isolated using cell surface markers for retinal therapy, cells selected from developing retinae by fluorescence-activated cell sorting based on Cd24a expression (using CD24 antibody) and/or Nt5e expression (using CD73 antibody) were transplanted into the wild-type or Crb1(rd8/rd8) or Prph2(rd2/rd2) mouse eye. The CD73/CD24-sorted cells migrated into the outer nuclear layer, acquired the morphology of mature photoreceptors and expressed outer segment markers. They showed an 18-fold higher integration efficiency than that of unsorted cells and 2.3-fold higher than cells sorted based on a single genetic marker, NrlGFP, expression. These proof-of-principle studies show that transplantation competent photoreceptor precursor cells can be efficiently isolated from a heterogeneous mix of cells using cell surface antigens without loss of viability for the purpose of retinal stem cell therapy. Refinement of the selection of donorphotoreceptor precursor cells can increase the number of integrated photoreceptor cells,which is a prerequisite for the restoration of sight.
Collapse
Affiliation(s)
- J Lakowski
- UCL Institute of Child Health, UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Shinoe T, Kuribayashi H, Saya H, Seiki M, Aburatani H, Watanabe S. Identification of CD44 as a cell surface marker for Müller glia precursor cells. J Neurochem 2010; 115:1633-42. [PMID: 20969572 DOI: 10.1111/j.1471-4159.2010.07072.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In the retina, both neurons and glia differentiate from a common progenitor population. CD44 cell surface antigen is a hyaluronic acid receptor expressed on mature Müller glial cells. We found that in the developing mouse retina, expression of CD44 was transiently observed at or around birth in a subpopulation of c-kit-positive retinal progenitor cells. During in vitro culture, purified CD44/c-kit-positive retinal progenitor cells exclusively differentiated into Müller glial cells and not into neurons, suggesting that CD44 marks a subpopulation of retinal progenitor cells that are fated to become glia. Over-expression of CD44 inhibited the extension of processes by Müller glial cells and neurons. Notch signaling is known to be involved in the specification of retinal progenitors into a glial fate. Activation of Notch signaling increased the number of CD44-positive cells, and treatment with the Notch signal inhibitor, DAPT, at early, but not later, stages of retinal development abolished both CD44-positive cells and Müller glial cells. Together, CD44 was identified as an early cell surface marker of the Müller glia lineage, and Notch signalling was involved in commitment of retinal progenitor cells to CD44 positive Müller glial precursor cells.
Collapse
Affiliation(s)
- Toru Shinoe
- Division of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
31
|
Liu Y, Huang M, Zhang Y, Li H, Xiao L, Liu J, Yuan B, Qin M, Li C, Yang M, Cai W. Screening genes related to development and injury of the mouse optic nerve by cDNA microarrays. Cell Mol Neurobiol 2010; 30:869-76. [PMID: 20336483 DOI: 10.1007/s10571-010-9515-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Accepted: 03/10/2010] [Indexed: 11/29/2022]
Abstract
The aim of this study was to screen genes related to the development and injury of the mouse optic nerve so as to provide possible target genes for gene-engineering therapy of central nervous system (CNS) injury. Gene expression was profiled by cDNA microarrays in the mouse superior colliculus at 8-time points during the development or following injury of the optic nerve; consequently, 1,095 highly expressed genes (ratio > or =2) were identified. Then, these genes were categorized functionally; there were 561 genes (51.19%) with unidentified functions and 534 genes (48.81%) with identified or partially identified functions. After discounting the overlapping genes, 486 genes with identified or partially identified functions were categorized into 17 functional groups. The 17 functional groups were as follows: I transcription regulation, II signal transduction, III protein synthesis, IV materials transporting, V RNA processing, VI metabolism-related genes, VII cell cycle or apoptosis-related genes, VIII extracellular matrix, IX protein folding and degradation, X cytoskeleton, XI histone metabolism, XII nervous system specific functional genes, XIII tumor related genes, XIV DNA replication and repair, XV axon growth and guidance, XVI immune response, and XVII cell adhesion. These genes may play key roles in the development, injury, and repairment of the optic nerve.
Collapse
Affiliation(s)
- Yunlai Liu
- Department of Histology & Embryology, The Third Military Medical University, Chongqing, 400038, China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Oron-Karni V, Farhy C, Elgart M, Marquardt T, Remizova L, Yaron O, Xie Q, Cvekl A, Ashery-Padan R. Dual requirement for Pax6 in retinal progenitor cells. Development 2008; 135:4037-4047. [PMID: 19004853 DOI: 10.1242/dev.028308] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Throughout the developing central nervous system, pre-patterning of the ventricular zone into discrete neural progenitor domains is one of the predominant strategies used to produce neuronal diversity in a spatially coordinated manner. In the retina, neurogenesis proceeds in an intricate chronological and spatial sequence, yet it remains unclear whether retinal progenitor cells (RPCs) display intrinsic heterogeneity at any given time point. Here, we performed a detailed study of RPC fate upon temporally and spatially confined inactivation of Pax6. Timed genetic removal of Pax6 appeared to unmask a cryptic divergence of RPCs into qualitatively divergent progenitor pools. In the more peripheral RPCs under normal circumstances, Pax6 seemed to prevent premature activation of a photoreceptor-differentiation pathway by suppressing expression of the transcription factor Crx. More centrally, Pax6 contributed to the execution of the comprehensive potential of RPCs: Pax6 ablation resulted in the exclusive generation of amacrine interneurons. Together, these data suggest an intricate dual role for Pax6 in retinal neurogenesis, while pointing to the cryptic divergence of RPCs into distinct progenitor pools.
Collapse
Affiliation(s)
- Varda Oron-Karni
- Sackler Faculty of Medicine, Human Molecular Genetics and Biochemistry, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| | - Chen Farhy
- Sackler Faculty of Medicine, Human Molecular Genetics and Biochemistry, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| | - Michael Elgart
- Sackler Faculty of Medicine, Human Molecular Genetics and Biochemistry, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| | - Till Marquardt
- European Neuroscience Institute, Developmental Neurobiology Laboratory, University of Göttingen Medical School/Max Planck Society, Grisebachstrasse 5, 37077 Göttingen, Germany
| | - Lena Remizova
- Sackler Faculty of Medicine, Human Molecular Genetics and Biochemistry, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| | - Orly Yaron
- Sackler Faculty of Medicine, Human Molecular Genetics and Biochemistry, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| | - Qing Xie
- Albert Einstein College of Medicine, Departments of Ophthalmology and Visual Sciences and Genetics, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Ales Cvekl
- Albert Einstein College of Medicine, Departments of Ophthalmology and Visual Sciences and Genetics, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Ruth Ashery-Padan
- Sackler Faculty of Medicine, Human Molecular Genetics and Biochemistry, Tel Aviv University, Ramat Aviv 69978, Tel Aviv, Israel
| |
Collapse
|
33
|
CD138/Syndecan-1 and SSEA-1 Mark Distinct Populations of Developing Ciliary Epithelium That Are Regulated Differentially by Wnt Signal. Stem Cells 2008; 26:3162-71. [DOI: 10.1634/stemcells.2008-0303] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
34
|
Perin L, Sedrakyan S, Da Sacco S, De Filippo R. Characterization of human amniotic fluid stem cells and their pluripotential capability. Methods Cell Biol 2008; 86:85-99. [PMID: 18442645 DOI: 10.1016/s0091-679x(08)00005-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Over the past decade, there has been ever-increasing emphasis placed on stem cells and their potential role in regenerative medicine for reconstruction of bio-artificial tissues and organs. Scientists have looked at various sources for pluripotential cells ranging from embryonic stem cells to adult stem cells. Amniocentesis is a well-established technique for the collection of cells derived from the human embryo. In this chapter, we are going to describe how to isolate, maintain in culture, and characterize the pluripotential capabilities of stem cells derived from amniocentesis in an in vitro and in vivo system. Cell samples are obtained from human pregnancies, and the progenitor cells are isolated from male fetuses with a normal karyotype in order to confirm the absence of maternal admixed cells. Progenitor cells express embryonic-specific cell markers, they show a high self-renewal capacity with 350 population doublings, and normal ploidy is confirmed by cell-cycle analyses. They maintain their undifferentiated state, pluripotential ability, clonogenicity, and telomere length over the population doublings. The progenitor cells are inducible to different cell lineages (osteogenic, adipogenic, skeletal muscle, endothelial, neuronal, and hepatic cells) under specific growth conditions. The ability to induce cell-type-specific differentiation is confirmed by phenotypic changes, immunocytochemistry, gene expression, and functional analyses. In addition, we will describe an application of these cells in an ex vivo and in vivo system for potential in organ (renal) regeneration. The progenitor cells described in this chapter have a high potential for expansion, and may be a good source for research and therapeutic applications where large numbers of cells are needed. Progenitor cells isolated during gestation may be beneficial for fetuses diagnosed with malformations and could be cryopreserved for future self-use.
Collapse
Affiliation(s)
- Laura Perin
- Childrens Hospital Los Angeles, Saban Research Institute, Developmental Biology Program, Keck School of Medicine, University of Southern California, USA
| | | | | | | |
Collapse
|
35
|
Hasegawa T, McLeod DS, Prow T, Merges C, Grebe R, Lutty GA. Vascular precursors in developing human retina. Invest Ophthalmol Vis Sci 2008; 49:2178-92. [PMID: 18436851 DOI: 10.1167/iovs.07-0632] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Prior investigation has demonstrated that angioblasts are present in the inner retinas of human embryos and fetuses and that they differentiate and organize to form the primordial retinal vasculature. The purpose of this study was to characterize these angioblasts further and examine ligands that might control their migration and differentiation. METHODS Immunohistochemistry was used to localize stroma-derived factor-1 (SDF-1), its receptor CXCR4, stem cell factor (SCF), and its receptor c-Kit on sections obtained from human eyes at from 6 to 23 weeks' gestation (WG). Coexpression of CD39 (marker for retinal angioblasts and endothelial cells) and CXCR4 or c-Kit was investigated by confocal microscopy. RESULTS SDF-1 was prominent in inner retina with the greatest reaction product near the internal limiting membrane (ILM). SCF immunoreactivity was also confined to the inner retina and increased significantly between 7 and 12 WG. The level of both ligands declined by 22 WG. A layer of CXCR4(+) and c-Kit(+) precursors, some of which coexpressed CD39, existed in the inner retina from 7 to 12 WG. With migration, c-Kit was downregulated, whereas CD39(+) cells continued to express CXCR4 as they formed cords. With canalization, CXCR4 expression was downregulated. CONCLUSIONS Embryonic human retina has a pool of precursors (CXCR4(+) and c-Kit(+)) that enlarged centrifugally during fetal development. From this pool emerges angioblasts, which migrate anteriorly into the nerve fiber layer where SDF-1 and SCF levels are highest. c-Kit expression declines with apparent migration, and CXCR4 expression declines with canalization of new vessels. Both SCF and SDF-1 are associated with the differentiation of retinal precursors into angioblasts and their migration to sites of vessel assembly.
Collapse
Affiliation(s)
- Takuya Hasegawa
- Wilmer Ophthalmological Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | | | | | | | | |
Collapse
|
36
|
Zhao J, Izumi T, Nunomura K, Satoh S, Watanabe S. MARCKS-like protein, a membrane protein identified for its expression in developing neural retina, plays a role in regulating retinal cell proliferation. Biochem J 2007; 408:51-9. [PMID: 17688421 PMCID: PMC2049077 DOI: 10.1042/bj20070826] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Membrane proteins are expressed in a specific manner in developing tissues, and characterization of these proteins is valuable because it allows them to be used as cell surface markers. Furthermore, they are potentially important for the regulation of organogenesis because some may participate in signal transduction. In the present study, we used proteomics to examine the comprehensive protein expression profile of the membrane fraction in the embryonic and adult mouse retina. We purified the retinal membrane fraction by sucrose-density-gradient centrifugation and analysed total proteins using shotgun analysis on a nanoflow LC-MS/MS (liquid chromatography tandem MS) system. Approximately half of the 326 proteins from the adult retina and a quarter of the 310 proteins from the embryonic retina (day 17) appeared to be membrane-associated proteins. Among these, MLP [MARCKS (myristoylated alanine-rich C-kinase substrate)-like protein], which shares approx. 50% amino acid identity with MARCKS, was selected for further characterization. The mRNA and surface protein expression of MLP decreased as retinal development progressed. Overexpression of MLP by retrovirus-mediated gene transfer enhanced the proliferation of retinal progenitor cells without affecting differentiation or cell migration in a retinal explant culture system. In contrast, MLP overexpression did not promote proliferation in fibroblasts (NIH 3T3 cells). Mutation analysis of MLP demonstrated that myristoylation was necessary to promote proliferation and that phosphorylation inhibited proliferation, indicating the functional importance of membrane localization.
Collapse
Affiliation(s)
- Jing Zhao
- *Department of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, 108-8639, Tokyo, Japan
| | - Tomonori Izumi
- †Department of Functional Proteomics, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, 108-8639 Tokyo, Japan
| | - Kazuto Nunomura
- †Department of Functional Proteomics, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, 108-8639 Tokyo, Japan
| | - Shinya Satoh
- *Department of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, 108-8639, Tokyo, Japan
| | - Sumiko Watanabe
- *Department of Molecular and Developmental Biology, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, 108-8639, Tokyo, Japan
- To whom correspondence should be addressed (email )
| |
Collapse
|
37
|
Kedees MH, Guz Y, Vuguin PM, Vargas C, Cui L, Steiner DF, Charron MJ, Teitelman G. Nestin expression in pancreatic endocrine and exocrine cells of mice lacking glucagon signaling. Dev Dyn 2007; 236:1126-33. [PMID: 17366624 PMCID: PMC4287279 DOI: 10.1002/dvdy.21112] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Nestin, a marker of neural stem cells, is also expressed by cells located in the epithelium of the pancreatic primordium and by a subpopulation of exocrine cells but not by endocrine cells. These findings raised the possibility that the pancreatic epithelium is heterogeneous and comprised of subpopulations of exocrine/nestin-positive and endocrine/nestin-negative precursor cells. We examined this issue in two mutant mouse models characterized by protracted expression of several embryonal properties in islet cells. One mutant line comprises mice lacking mature glucagon due to abrogation of proprotein convertase-2 (PC2(-/-)), responsible for the conversion of proglucagon into glucagon, while the second line consists of mice with a global deletion of the glucagon receptor (Gcgr(-/-)). We demonstrate that nestin is transiently expressed by acinar cells and by insulin and glucagon cells of islets of both lines of mice. In addition, the lack of glucagon signaling increased nestin mRNA levels in pancreas of mutant embryos and adult mice. We conclude that nestin+ cells located in the pancreatic primordium generate the cells of the endocrine and exocrine lineages. Furthermore, our results suggest that nestin expression is regulated by glucagon signaling.
Collapse
Affiliation(s)
- Mamdouh H. Kedees
- Department of Anatomy and Cell Biology, SUNY-Downstate Medical Center, Brooklyn, New York
| | - Yelena Guz
- Department of Anatomy and Cell Biology, SUNY-Downstate Medical Center, Brooklyn, New York
| | - Patricia M. Vuguin
- Department of Pediatrics, Albert Einstein College of Medicine, Bronx, New York
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
| | - Carlos Vargas
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
| | - Lingguang Cui
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
| | - Donald F. Steiner
- Department of Biochemistry, University of Chicago, Chicago, Illinois
| | - Maureen J. Charron
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
| | - Gladys Teitelman
- Department of Anatomy and Cell Biology, SUNY-Downstate Medical Center, Brooklyn, New York
| |
Collapse
|