1
|
Balzamino BO, Cacciamani A, Dinice L, Cecere M, Pesci FR, Ripandelli G, Micera A. Retinal Inflammation and Reactive Müller Cells: Neurotrophins' Release and Neuroprotective Strategies. BIOLOGY 2024; 13:1030. [PMID: 39765697 PMCID: PMC11673524 DOI: 10.3390/biology13121030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025]
Abstract
Millions of people worldwide suffer from retinal disorders. Retinal diseases require prompt attention to restore function or reduce progressive impairments. Genetics, epigenetics, life-styling/quality and external environmental factors may contribute to developing retinal diseases. In the physiological retina, some glial cell types sustain neuron activities by guaranteeing ion homeostasis and allowing effective interaction in synaptic transmission. Upon insults, glial cells interact with neuronal and the other non-neuronal retinal cells, at least in part counteracting the biomolecular changes that may trigger retinal complications and vision loss. Several epigenetic and oxidative stress mechanisms are quickly activated to release factors that in concert with growth, fibrogenic and angiogenic factors can influence the overall microenvironment and cell-to-cell response. Reactive Müller cells participate by secreting neurotrophic/growth/angiogenic factors, cytokines/chemokines, cytotoxic/stress molecules and neurogenic inflammation peptides. Any attempt to maintain/restore the physiological condition can be interrupted by perpetuating insults, vascular dysfunction and neurodegeneration. Herein, we critically revise the current knowledge on the cell-to-cell and cell-to-mediator interplay between Müller cells, astrocytes and microglia, with respect to pro-con modulators and neuroprotective/detrimental activities, as observed by using experimental models or analyzing ocular fluids, altogether contributing a new point of view to the field of research on precision medicine.
Collapse
Affiliation(s)
- Bijorn Omar Balzamino
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Science, IRCCS-Fondazione Bietti, via di Santo Stefano Rotondo 6, 00184 Rome, Italy; (B.O.B.); (L.D.)
| | - Andrea Cacciamani
- Surgical Retina Research Unit, IRCCS-Fondazione Bietti, via di Santo Stefano Rotondo 6, 00184 Rome, Italy; (A.C.); (M.C.); (F.R.P.); (G.R.)
| | - Lucia Dinice
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Science, IRCCS-Fondazione Bietti, via di Santo Stefano Rotondo 6, 00184 Rome, Italy; (B.O.B.); (L.D.)
| | - Michela Cecere
- Surgical Retina Research Unit, IRCCS-Fondazione Bietti, via di Santo Stefano Rotondo 6, 00184 Rome, Italy; (A.C.); (M.C.); (F.R.P.); (G.R.)
| | - Francesca Romana Pesci
- Surgical Retina Research Unit, IRCCS-Fondazione Bietti, via di Santo Stefano Rotondo 6, 00184 Rome, Italy; (A.C.); (M.C.); (F.R.P.); (G.R.)
| | - Guido Ripandelli
- Surgical Retina Research Unit, IRCCS-Fondazione Bietti, via di Santo Stefano Rotondo 6, 00184 Rome, Italy; (A.C.); (M.C.); (F.R.P.); (G.R.)
| | - Alessandra Micera
- Research and Development Laboratory for Biochemical, Molecular and Cellular Applications in Ophthalmological Science, IRCCS-Fondazione Bietti, via di Santo Stefano Rotondo 6, 00184 Rome, Italy; (B.O.B.); (L.D.)
| |
Collapse
|
2
|
Age- and cell cycle-related expression patterns of transcription factors and cell cycle regulators in Müller glia. Sci Rep 2022; 12:19584. [PMID: 36379991 PMCID: PMC9666513 DOI: 10.1038/s41598-022-23855-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Mammalian Müller glia express transcription factors and cell cycle regulators essential for the function of retinal progenitors, indicating the latent neurogenic capacity; however, the role of these regulators remains unclear. To gain insights into the role of these regulators in Müller glia, we analyzed expression of transcription factors (Pax6, Vsx2 and Nfia) and cell cycle regulators (cyclin D1 and D3) in rodent Müller glia, focusing on their age- and cell cycle-related expression patterns. Expression of Pax6, Vsx2, Nfia and cyclin D3, but not cyclin D1, increased in Müller glia during development. Photoreceptor injury induced cell cycle-associated increase of Vsx2 and cyclin D1, but not Pax6, Nfia, and cyclin D3. In dissociated cultures, cell cycle-associated increase of Pax6 and Vsx2 was observed in Müller glia from P10 mice but not from P21 mice. Nfia levels were highly correlated with EdU incorporation suggesting their activation during S phase progression. Cyclin D1 and D3 were transiently upregulated in G1 phase but downregulated after S phase entry. Our findings revealed previously unknown links between cell cycle progression and regulator protein expression, which likely affect the cell fate decision of proliferating Müller glia.
Collapse
|
3
|
Lee K, Choi JO, Hwang A, Bae HW, Kim CY. Ciliary Neurotrophic Factor Derived From Astrocytes Protects Retinal Ganglion Cells Through PI3K/AKT, JAK/STAT, and MAPK/ERK Pathways. Invest Ophthalmol Vis Sci 2022; 63:4. [PMID: 35925584 PMCID: PMC9363680 DOI: 10.1167/iovs.63.9.4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 07/11/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose The purpose of this study was to investigate the roles of ciliary neurotrophic factor (CNTF) on the protective effects of astrocytes on retinal ganglion cells (RGCs). Methods Primary RGCs were isolated from neonatal rats. Oxidative stress was induced, and the effects of co-culture with astrocytes and CNTF treatment on RGCs were evaluated. The pathways commonly altered by astrocytes and CNTF were investigated. Effects of each pathway were investigated using pathway inhibitors against PI3K/AKT, JAK/STAT, and MAPK/ERK. RNA sequencing was performed to identify the genes upregulated and downregulated by CNTF treatment. Results Astrocytes improved the viability and increased β3-tubulin expression in RGCs. The concentration of CNTF increased in the RGC-astrocyte co-culture medium. The protective effects of astrocytes were abolished by neutralization with the anti-CNTF antibody; thus, CNTF may play an important role in the effects mediated by astrocytes. Furthermore, CNTF treatment alone enhanced the viability and β3-tubulin expression of RGCs and increased the population of viable RGCs under oxidative stress. The PI3K/AKT pathway was associated with both RGC viability and β3-tubulin expression. However, the JAK/STAT pathway increased the viability of RGCs, whereas the MAPK/ERK pathway was associated with β3-tubulin expression. RNA sequencing revealed the CNTF-upregulated genes associated with response to DNA damage and downregulated genes associated with photoreceptor cell differentiation. Conclusions Our data revealed protective effects of astrocyte-derived CNTF on RGCs. In addition, we showed that multiple pathways exert these protective effects and identified the novel genes involved. These results may be helpful in developing treatments for RGC injury.
Collapse
Affiliation(s)
- Kwanghyun Lee
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Ophthalmology, National Health Insurance Service Ilsan Hospital, Goyang, Gyeonggi-do, Republic of Korea
| | - Jin-Ok Choi
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ahreum Hwang
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyoung Won Bae
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chan Yun Kim
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
4
|
Guimarães RPDM, Landeira BS, Coelho DM, Golbert DCF, Silveira MS, Linden R, de Melo Reis RA, Costa MR. Evidence of Müller Glia Conversion Into Retina Ganglion Cells Using Neurogenin2. Front Cell Neurosci 2018; 12:410. [PMID: 30483060 PMCID: PMC6240664 DOI: 10.3389/fncel.2018.00410] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 10/22/2018] [Indexed: 01/09/2023] Open
Abstract
Degenerative retinopathies are the leading causes of irreversible visual impairment in the elderly, affecting hundreds of millions of patients. Müller glia cells (MGC), the main type of glia found in the vertebrate retina, can resume proliferation in the rodent adult injured retina but contribute weakly to tissue repair when compared to zebrafish retina. However, postnatal and adult mouse MGC can be genetically reprogrammed through the expression of the transcription factor (TF) Achaete-scute homolog 1 (ASCL1) into induced neurons (iNs), displaying key hallmarks of photoreceptors, bipolar and amacrine cells, which may contribute to regenerate the damaged retina. Here, we show that the TF neurogenin 2 (NEUROG2) is also sufficient to lineage-reprogram postnatal mouse MGC into iNs. The efficiency of MGC lineage conversion by NEUROG2 is similar to that observed after expression of ASCL1 and both TFs induce the generation of functionally active iNs. Treatment of MGC cultures with EGF and FGF2 prior to Neurog2 or Ascl1 expression enhances reprogramming efficiencies, what can be at least partially explained by an increase in the frequency of MGCs expressing sex determining region Y (SRY)-box 2 (SOX2). Transduction of either Neurog2 or Ascl1 led to the upregulation of key retina neuronal genes in MGC-derived iNs, but only NEUROG2 induced a consistent increase in the expression of putative retinal ganglion cell (RGC) genes. Moreover, in vivo electroporation of Neurog2 in late progenitors from the neonatal rat retina, which are transcriptionally similar to MGCs, also induced a shift in the generation of retinal cell subtypes, favoring neuronal differentiation at the expense of MGCs and resuming the generation of RGCs. Altogether, our data indicate that NEUROG2 induces lineage conversion of postnatal rodent MGCs into RGC-like iNs in vitro and resumes the generation of this neuronal type from late progenitors of the retina in vivo.
Collapse
Affiliation(s)
- Roberta Pereira de Melo Guimarães
- Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil.,Lab Neurogenesis, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Lab Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Diego Marques Coelho
- Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil.,Bioinformatics Multidisciplinary Environment, IMD, Federal University of Rio Grande do Norte, Rio de Janeiro, Brazil
| | | | - Mariana S Silveira
- Lab Neurogenesis, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rafael Linden
- Lab Neurogenesis, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ricardo A de Melo Reis
- Lab Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos R Costa
- Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
5
|
Jindal N, Banik A, Prabhakar S, Vaiphie K, Anand A. Alteration of Neurotrophic Factors After Transplantation of Bone Marrow Derived Lin-ve Stem Cell in NMDA-Induced Mouse Model of Retinal Degeneration. J Cell Biochem 2017; 118:1699-1711. [PMID: 27935095 DOI: 10.1002/jcb.25827] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 12/06/2016] [Indexed: 02/02/2023]
Abstract
Retinal ganglion cell layer (RGCs) is one of the important layers of retina, depleted in Glaucoma. Loss of RGC neurons is a major cellular mechanism involved in its pathogenesis resulting in severe vision loss. Stem cell therapy has emerged as a potential strategy to arrest the apoptotic loss of RGCs and also replace the degenerative cells in damaged retina. Here, we have investigated the incorporation and survival of mouse bone marrow derived Lin-ve stem cells in N-methyl-d-aspartate (NMDA)-induced mouse model of retinal degeneration. Two days after intravitreal injection of NMDA (100 mM) showed significant decrease in ganglion cell number and increase in TUNEL positive apoptotic cells in retinal layers. The injury was further characterized by immunohistochemical expression of Brn3b, GFAP, Bcl2, pCREB, CNTF, GDNF, and BDNF in retinal layers. Lin-ve cells (100,000 dose) were intravitreally transplanted after 2 days of injury and evaluated after 7, 14, and 21 days of transplantation. Transplanted cells were found to have migrated from intravitreal space and incorporated into injured retina at 7, 14, and 21 days post-transplantation. At 21 days Brn3b, CNTF, and BDNF expression was found to be upregulated whereas GDNF was downregulated when compared to respective injury time points. Molecular data showed decrease in the expression of Brn3b, BDNF, CNTF, and GDNF post transplantation when compared with injury groups. This study reveals that Lin-ve stem cells may exert neuroprotective effect in damaged retina mediated by participation of neurotrophic factors induced by stem cell transplantation at the site of injury. J. Cell. Biochem. 118: 1699-1711, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Neeru Jindal
- Neuroscience Research Lab, Department of Neurology, PGIMER, Chandigarh 160012, India
| | - Avijit Banik
- Neuroscience Research Lab, Department of Neurology, PGIMER, Chandigarh 160012, India
| | - Sudesh Prabhakar
- Neuroscience Research Lab, Department of Neurology, PGIMER, Chandigarh 160012, India
| | - Kim Vaiphie
- Department of Histopathology, PGIMER, Chandigarh 160012, India
| | - Akshay Anand
- Neuroscience Research Lab, Department of Neurology, PGIMER, Chandigarh 160012, India
| |
Collapse
|
6
|
Wu ZK, Cao L, Zhang XY, Song WT, Xia XB. Promotion on the differentiation of retinal Müller cells into retinal ganglion cells by Brn-3b. Int J Ophthalmol 2016; 9:948-54. [PMID: 27500099 DOI: 10.18240/ijo.2016.07.03] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 11/24/2015] [Indexed: 11/23/2022] Open
Abstract
AIM To investigate the role of Brn-3b in differentiation process of stem cells derived from retinal Müller cells into the ganglion cell. METHODS The passage culture method of Müller cells from retina of newborn Sprague Dawley rats was carried out by repeated incomplete pancreatic enzyme digestion method. The cells were detected by fluorescence-activated cell sorter (FACS), immunohistochemistry technology and reverse transcription-polymerase chain reaction (RT-PCR) to determine the purity. The third passage of cells was induced in the serum-free dedifferentiation medium. The expression of the specific markers Ki-67 and nestin of retinal stem cells was measured by RT-PCR and Western blot. The cell proliferation of retinal stem cells was detected by 5-ethynyl-2'-deoxyuridine (Edu) staining. The cells were randomly divided into 5 groups as follows: group A: Brn-3bsiRNA group; group B: Brn-3b control siRNA group; group C: pGC-Brn-3b-green fluorescent protein (GFP) group; group D: pGC-GFP group; group E: control group (without any handling). The purified Müller cells were cultured for 3-7d, then, the percentage of ganglion cells was counted by immunofluorescence staining. RESULTS FACS demonstrated the purity of retinal Müller cells was more 97.44%. A few spherical cell spheres appeared. Immunofluorescence staining showed that stem cells within the spheres were positive for retinal stem cell-specific markers nestin (red fluorescence, 92.94%±6.48%) and Ki-67 (green fluorescence, 85.96%±6.04%). Meanwhile, RT-PCR analysis showed cell spheres in the culture to have expressed a battery of transcripts characteristic of stem cells such as nestin and Ki-67, which were absent in the Müller cells. Western blot analysis further confirmed the expression of nestin and Ki-67 in the cell spheres but not in the Müller cells. Edu staining showed most of the nuclei within the cell spheres were stained red (82.80%±6.65%), suggesting the new cell spheres had the capacity for effective proliferation. The statistics result showed the difference between Brn-3bsiRNA group and Brn-3b control siRNA group or the control group was significant (F=15, P<0.05), while the difference between Brn-3b control siRNA group or the control group was not statistically significant (P>0.05). CONCLUSION The repeated incomplete pancreatic enzyme digestion method is an efficient and practical method to purify retinal Müller cells. Retinal stem cells were successfully cloned in the dedifferentiational medium. Retinal Müller cells are accessible sources of retinal stem cells. Brn-3b is an important regulatory gene in stem cells differentiated into retinal ganglion cell.
Collapse
Affiliation(s)
- Zhen-Kai Wu
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Lan Cao
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Xue-Yong Zhang
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Wei-Tao Song
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Xiao-Bo Xia
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|
7
|
Corso-Díaz X, Simpson EM. Nr2e1 regulates retinal lamination and the development of Müller glia, S-cones, and glycineric amacrine cells during retinogenesis. Mol Brain 2015; 8:37. [PMID: 26092486 PMCID: PMC4475312 DOI: 10.1186/s13041-015-0126-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2015] [Accepted: 05/23/2015] [Indexed: 12/25/2022] Open
Abstract
Background Nr2e1 is a nuclear receptor crucial for neural stem cell proliferation and maintenance. In the retina, lack of Nr2e1 results in premature neurogenesis, aberrant blood vessel formation and dystrophy. However, the specific role of Nr2e1 in the development of different retinal cell types and its cell-autonomous and non-cell autonomous function(s) during eye development are poorly understood. Results Here, we studied the retinas of P7 and P21 Nr2e1frc/frc mice and Nr2e1+/+ ↔ Nr2e1frc/frc chimeras. We hypothesized that Nr2e1 differentially regulates the development of various retinal cell types, and thus the cellular composition of Nr2e1frc/frc retinas does not simply reflect an overrepresentation of cells born early and underrepresentation of cells born later as a consequence of premature neurogenesis. In agreement with our hypothesis, lack of Nr2e1 resulted in increased numbers of glycinergic amacrine cells with no apparent increase in other amacrine sub-types, normal numbers of Müller glia, the last cell-type to be generated, and increased numbers of Nr2e1frc/frc S-cones in chimeras. Furthermore, Nr2e1frc/frc Müller glia were mispositioned in the retina and misexpressed the ganglion cell-specific transcription factor Brn3a. Nr2e1frc/frc retinas also displayed lamination defects including an ectopic neuropil forming an additional inner plexiform layer. In chimeric mice, retinal thickness was rescued by 34 % of wild-type cells and Nr2e1frc/frc dystrophy-related phenotypes were no longer evident. However, the formation of an ectopic neuropil, misexpression of Brn3a in Müller glia, and abnormal cell numbers in the inner and outer nuclear layers at P7 were not rescued by wild-type cells. Conclusions Together, these results show that Nr2e1, in addition to having a role in preventing premature cell cycle exit, participates in several other developmental processes during retinogenesis including neurite organization in the inner retina and development of glycinergic amacrine cells, S-cones, and Müller glia. Nr2e1 also regulates various aspects of Müller glia differentiation cell-autonomously. However, Nr2e1 does not have a cell-autonomous role in preventing retinal dystrophy. Thus, Nr2e1 regulates processes involved in neurite development and terminal retinal cell differentiation. Electronic supplementary material The online version of this article (doi:10.1186/s13041-015-0126-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ximena Corso-Díaz
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, 950 W 28 Ave, Vancouver, V5Z 4H4, BC, Canada.,Genetics Graduate Program, University of British Columbia, Vancouver, V6T 1Z2, BC, Canada
| | - Elizabeth M Simpson
- Centre for Molecular Medicine and Therapeutics at the Child and Family Research Institute, University of British Columbia, 950 W 28 Ave, Vancouver, V5Z 4H4, BC, Canada. .,Genetics Graduate Program, University of British Columbia, Vancouver, V6T 1Z2, BC, Canada. .,Department of Medical Genetics, University of British Columbia, Vancouver, V6T 1Z3, BC, Canada. .,Department of Psychiatry, University of British Columbia, Vancouver, V6T 2A1, BC, Canada.
| |
Collapse
|
8
|
|
9
|
Yip HK. Retinal stem cells and regeneration of vision system. Anat Rec (Hoboken) 2013; 297:137-60. [PMID: 24293400 DOI: 10.1002/ar.22800] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 09/13/2013] [Indexed: 12/14/2022]
Abstract
The vertebrate retina is a well-characterized model for studying neurogenesis. Retinal neurons and glia are generated in a conserved order from a pool of mutlipotent progenitor cells. During retinal development, retinal stem/progenitor cells (RPC) change their competency over time under the influence of intrinsic (such as transcriptional factors) and extrinsic factors (such as growth factors). In this review, we summarize the roles of these factors, together with the understanding of the signaling pathways that regulate eye development. The information about the interactions between intrinsic and extrinsic factors for retinal cell fate specification is useful to regenerate specific retinal neurons from RPCs. Recent studies have identified RPCs in the retina, which may have important implications in health and disease. Despite the recent advances in stem cell biology, our understanding of many aspects of RPCs in the eye remains limited. PRCs are present in the developing eye of all vertebrates and remain active in lower vertebrates throughout life. In mammals, however, PRCs are quiescent and exhibit very little activity and thus have low capacity for retinal regeneration. A number of different cellular sources of RPCs have been identified in the vertebrate retina. These include PRCs at the retinal margin, pigmented cells in the ciliary body, iris, and retinal pigment epithelium, and Müller cells within the retina. Because PRCs can be isolated and expanded from immature and mature eyes, it is possible now to study these cells in culture and after transplantation in the degenerated retinal tissue. We also examine current knowledge of intrinsic RPCs, and human embryonic stems and induced pluripotent stem cells as potential sources for cell transplant therapy to regenerate the diseased retina.
Collapse
Affiliation(s)
- Henry K Yip
- Department of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Adminstrative Region, People's Republic of China; Research Center of Heart, Brain, Hormone and Healthy Aging, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Adminstrative Region, People's Republic of China; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Pokfulam, Hong Kong Special Adminstrative Region, People's Republic of China
| |
Collapse
|
10
|
Glutamate-induced epigenetic and morphological changes allow rat Müller cell dedifferentiation but not further acquisition of a photoreceptor phenotype. Neuroscience 2013; 254:347-60. [DOI: 10.1016/j.neuroscience.2013.09.048] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 09/23/2013] [Accepted: 09/25/2013] [Indexed: 11/22/2022]
|
11
|
Stutz B, da Conceição FSL, Santos LE, Cadilhe DV, Fleming RL, Acquarone M, Gardino PF, de Melo Reis RA, Dickson PW, Dunkley PR, Rehen S, Houzel JC, de Mello FG. Murine dopaminergic Müller cells restore motor function in a model of Parkinson's disease. J Neurochem 2013; 128:829-40. [DOI: 10.1111/jnc.12475] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 09/27/2013] [Accepted: 09/27/2013] [Indexed: 11/30/2022]
Affiliation(s)
- Bernardo Stutz
- Instituto de Biofísica Carlos Chagas Filho; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
| | | | - Luís Eduardo Santos
- Instituto de Biofísica Carlos Chagas Filho; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
| | - Daniel Veloso Cadilhe
- Instituto de Ciências Biomédicas; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
| | - Renata L. Fleming
- Instituto de Biofísica Carlos Chagas Filho; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
| | - Mariana Acquarone
- Instituto de Ciências Biomédicas; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
| | - Patrícia F. Gardino
- Instituto de Biofísica Carlos Chagas Filho; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
| | - Ricardo A. de Melo Reis
- Instituto de Biofísica Carlos Chagas Filho; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
| | - Phillip W. Dickson
- School of Biomedical Sciences and Pharmacy; University of Newcastle; Callaghan NSW Australia
| | - Peter R. Dunkley
- School of Biomedical Sciences and Pharmacy; University of Newcastle; Callaghan NSW Australia
| | - Stevens Rehen
- Instituto de Ciências Biomédicas; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
| | - Jean-Christophe Houzel
- Instituto de Ciências Biomédicas; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
| | - Fernando G. de Mello
- Instituto de Biofísica Carlos Chagas Filho; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ Brazil
| |
Collapse
|
12
|
Pollak J, Wilken MS, Ueki Y, Cox KE, Sullivan JM, Taylor RJ, Levine EM, Reh TA. ASCL1 reprograms mouse Muller glia into neurogenic retinal progenitors. Development 2013; 140:2619-31. [PMID: 23637330 PMCID: PMC3666387 DOI: 10.1242/dev.091355] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2013] [Indexed: 12/14/2022]
Abstract
Non-mammalian vertebrates have a robust ability to regenerate injured retinal neurons from Müller glia (MG) that activate the gene encoding the proneural factor Achaete-scute homolog 1 (Ascl1; also known as Mash1 in mammals) and de-differentiate into progenitor cells. By contrast, mammalian MG have a limited regenerative response and fail to upregulate Ascl1 after injury. To test whether ASCL1 could restore neurogenic potential to mammalian MG, we overexpressed ASCL1 in dissociated mouse MG cultures and intact retinal explants. ASCL1-infected MG upregulated retinal progenitor-specific genes and downregulated glial genes. Furthermore, ASCL1 remodeled the chromatin at its targets from a repressive to an active configuration. MG-derived progenitors differentiated into cells that exhibited neuronal morphologies, expressed retinal subtype-specific neuronal markers and displayed neuron-like physiological responses. These results indicate that a single transcription factor, ASCL1, can induce a neurogenic state in mature MG.
Collapse
Affiliation(s)
- Julia Pollak
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
- Neurobiology and Behavior Program, University of Washington, Seattle, WA 98195, USA
| | - Matthew S. Wilken
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Yumi Ueki
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Kristen E. Cox
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Jane M. Sullivan
- Neurobiology and Behavior Program, University of Washington, Seattle, WA 98195, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA
| | - Russell J. Taylor
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Edward M. Levine
- Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah, Salt Lake City, UT 84132, USA
| | - Thomas A. Reh
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
- Neurobiology and Behavior Program, University of Washington, Seattle, WA 98195, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
13
|
Müller glia express rhodopsin in a mouse model of inherited retinal degeneration. Neuroscience 2012; 225:152-61. [DOI: 10.1016/j.neuroscience.2012.08.066] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 08/20/2012] [Accepted: 08/30/2012] [Indexed: 11/22/2022]
|
14
|
Katsman D, Stackpole EJ, Domin DR, Farber DB. Embryonic stem cell-derived microvesicles induce gene expression changes in Müller cells of the retina. PLoS One 2012; 7:e50417. [PMID: 23226281 PMCID: PMC3511553 DOI: 10.1371/journal.pone.0050417] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 10/19/2012] [Indexed: 12/20/2022] Open
Abstract
Cell-derived microvesicles (MVs), recognized as important components of cell-cell communication, contain mRNAs, miRNAs, proteins and lipids and transfer their bioactive contents from parent cells to cells of other origins. We have studied the effect that MVs released from embryonic stem cells (ESMVs) have on retinal progenitor Müller cells. Cultured human Müller cells were exposed to mouse ESMVs every 48 hours for a total of 9 treatments. Morphological changes were observed by light microscopy in the treated cells, which grew as individual heterogeneous cells, compared to the uniform, spindle-like adherent cellular sheets of untreated cells. ESMVs transferred to Müller cells embryonic stem cell (ESC) mRNAs involved in the maintenance of pluripotency, including Oct4 and Sox2, and the miRNAs of the 290 cluster, important regulators of the ESC-specific cell cycle. Moreover, ESMV exposure induced up-regulation of the basal levels of endogenous human Oct4 mRNA in Müller cells. mRNA and miRNA microarrays of ESMV-treated vs. untreated Müller cells revealed the up-regulation of genes and miRNAs involved in the induction of pluripotency, cellular proliferation, early ocular genes and genes important for retinal protection and remodeling, as well as the down-regulation of inhibitory and scar-related genes and miRNAs involved in differentiation and cell cycle arrest. To further characterize the heterogeneous cell population of ESMV-treated Müller cells, their expression of retinal cell markers was compared to that in untreated control cells by immunocytochemistry. Markers for amacrine, ganglion and rod photoreceptors were present in treated but not in control Müller cells. Together, our findings indicate that ESMs induce de-differentiation and pluripotency in their target Müller cells, which may turn on an early retinogenic program of differentiation.
Collapse
Affiliation(s)
- Diana Katsman
- Jules Stein Eye Institute and Department of Ophthalmology, University of California Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
| | - Emma J. Stackpole
- Jules Stein Eye Institute and Department of Ophthalmology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Daniel R. Domin
- Jules Stein Eye Institute and Department of Ophthalmology, University of California Los Angeles, Los Angeles, California, United States of America
| | - Debora B. Farber
- Jules Stein Eye Institute and Department of Ophthalmology, University of California Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
- Brain Research Institute, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
15
|
Wohl SG, Schmeer CW, Isenmann S. Neurogenic potential of stem/progenitor-like cells in the adult mammalian eye. Prog Retin Eye Res 2012; 31:213-42. [DOI: 10.1016/j.preteyeres.2012.02.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 02/04/2012] [Accepted: 02/06/2012] [Indexed: 11/26/2022]
|
16
|
Wang Z, Sugano E, Isago H, Murayama N, Tamai M, Tomita H. Notch signaling pathway regulates proliferation and differentiation of immortalized Müller cells under hypoxic conditions in vitro. Neuroscience 2012; 214:171-80. [PMID: 22525134 DOI: 10.1016/j.neuroscience.2012.04.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Revised: 03/26/2012] [Accepted: 04/12/2012] [Indexed: 12/14/2022]
Abstract
Previous studies have indicated that Müller glia in chick and fish retinas can re-enter the cell cycle, express progenitor genes, and regenerate neurons via the Notch signaling pathway in response to retinal damage or growth factors. Here, we investigated the role of Notch signaling and the effect of hypoxia, as a means to induce retinal damage, on the proliferation of an immortalized Müller cell line (rMC-1 cells). Our data showed that rMC-1 cells expressed Müller glia and neural and retinal progenitor markers but did not express neuronal or retinal markers. Hypoxia increased rMC-1 cell proliferation by activating the positive cell-cycle regulators, cyclins A and D1, as well as the neural and retinal progenitor markers, Notch1, Hes1, nestin, Sox2, Msi1, Pax6, and NeuroD1. However, hypoxia did not significantly influence the expression of Müller glial markers GS, CRALBP, and cyclin D3 or the death of the rMC-1 cells. The increase in cell proliferation induced by hypoxia was greatly attenuated by blocking Notch signaling with the inhibitor DAPT, resulting in the reduced expression of positive cell-cycle regulators (cyclins A and D1) and neural and retinal progenitor markers (Notch1, Hes1, Sox2, Pax6, and NeuroD1). Blockade of the Notch signaling pathway by DAPT after hypoxia promoted the differentiation of rMC-1 cells to neurons, as demonstrated by the induction of neural marker (Tuj1), retinal amacrine (Syntaxin1), and retinal ganglion cell (Brn3b) markers, although the expression of the latter marker was low. Taken together, our data indicate that Notch signaling is required for proliferation under hypoxic conditions either by activating the positive cell-cycle regulators or by skewing their de-differentiation towards a neural progenitor lineage. These findings indicate that the Notch signaling pathway regulates hypoxia-induced proliferation and differentiation of Müller glia.
Collapse
Affiliation(s)
- Z Wang
- Tohoku University Institute for International Advanced Interdisciplinary Research, 4-1 Seiryo-machi, Aoba-ku, Sendai, Japan
| | | | | | | | | | | |
Collapse
|
17
|
Liu Y, Liu RR, Wang L, Zeng L, Long ZY, Wu YM. The effects of different phenotype astrocytes on neural stem cells differentiation in co-culture. Neurosci Lett 2011; 508:61-6. [PMID: 22206833 DOI: 10.1016/j.neulet.2011.12.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 12/01/2011] [Accepted: 12/13/2011] [Indexed: 01/09/2023]
Abstract
Astrocytes were reported to show neuroprotective effects on neurons, but there was no direct evidence for a functional relationship between astrocytes and neural stem cells (NSCs). In this experiments, we examined neuronal differentiation of NSCs induced by protoplasmic and fibrous astrocytes in a co-culture model respectively. Two types of astrocytes and NSCs were isolated from E13 to 15 cortex of rats. The neuronal differentiation of NSCs was examined after co-culture with two kinds of astrocytes. There were more neuronal marker β-tublin III positive cells from NSCs co-cultured with protoplasmic astrocytes. However the differentiated neurons, whether co-cultured with protoplasmic astrocytes or fibrous astrocytes, both expressed glutamate AMPA receptor subunit GluR2 protein and exhibited biological electrical reactivity after stimulated by glutamine. Therefore, these findings indicated that two types of astrocytes could induce the differentiation of NSCs and also possibly induce functional maturation of differentiated neurons, among which protoplasmic astrocytes have the ability to promote neuronal differentiation of NSCs compared with fibrous astrocytes.
Collapse
Affiliation(s)
- Yuan Liu
- 3rd Department of Research Institute of Surgery, Daping Hospital, The Third Military Medical University, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing 400042, China
| | | | | | | | | | | |
Collapse
|
18
|
Simón MV, De Genaro P, Abrahan CE, de los Santos B, Rotstein NP, Politi LE. Müller glial cells induce stem cell properties in retinal progenitors in vitro and promote their further differentiation into photoreceptors. J Neurosci Res 2011; 90:407-21. [DOI: 10.1002/jnr.22747] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 06/18/2011] [Accepted: 06/21/2011] [Indexed: 01/12/2023]
|
19
|
Nasonkin IO, Lazo K, Hambright D, Brooks M, Fariss R, Swaroop A. Distinct nuclear localization patterns of DNA methyltransferases in developing and mature mammalian retina. J Comp Neurol 2011; 519:1914-30. [PMID: 21452232 PMCID: PMC12058000 DOI: 10.1002/cne.22613] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
DNA methyltransferases--DNMT1, DNMT3a, and DNMT3b--produce methylation patterns that dynamically regulate chromatin remodeling and gene expression. The vertebrate retina provides an ideal model to elucidate molecular control of neurogenesis as all neuronal cell types and Müller glia are generated in a conserved order from common pools of progenitor cells. As a prelude to exploring epigenetic regulation of mammalian retinal development, we investigated the expression of Dnmt1, Dnmt3a, and Dnmt3b in the mouse retina from embryonic day (E) 10.5 to 10 months of age. High levels of transcripts for all three Dnmt genes were observed in early stages of retinal differentiation, with significantly reduced expression after birth. Although DNMT1 protein is abundant in retinal progenitors at E10.5, it becomes restricted to postmitotic cells by E15.5. Most cells in the postnatal retina show nuclear immunostaining of DNMT1; however, the photoreceptors exhibit distinctive patterns. In rods, weak expression of DNMT1 is detected in perinuclear region and in the nucleus, whereas a strong nuclear labeling is evident in cones. DNMT3a and DNMT3b show a discrete pattern in developing retina with high expression at E11.5, little or no immunostaining by E15.5, and then postnatal expression overlapping with DNMT1 in early born neurons (ganglion, amacrine and horizontal cells, and cones). Robust nuclear localization of DNMTs in cones compared to rods suggests a potential role of DNA methylation in differential remodeling of chromatin in these two specialized neurons. Our studies indicate that DNA methyltransferases contribute to the establishment and maturation of cell fates during retinal development.
Collapse
Affiliation(s)
- Igor O. Nasonkin
- Neurobiology-Neurodegeneration & Repair Laboratory (N-NRL), National Institutes of Health, Bethesda, Maryland 20892
| | - Kevin Lazo
- Neurobiology-Neurodegeneration & Repair Laboratory (N-NRL), National Institutes of Health, Bethesda, Maryland 20892
| | - Dustin Hambright
- Neurobiology-Neurodegeneration & Repair Laboratory (N-NRL), National Institutes of Health, Bethesda, Maryland 20892
| | - Matthew Brooks
- Neurobiology-Neurodegeneration & Repair Laboratory (N-NRL), National Institutes of Health, Bethesda, Maryland 20892
| | - Robert Fariss
- Biological Imaging Core, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Anand Swaroop
- Neurobiology-Neurodegeneration & Repair Laboratory (N-NRL), National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
20
|
Changes in Musashi-1 subcellular localization correlate with cell cycle exit during postnatal retinal development. Exp Eye Res 2011; 92:344-52. [DOI: 10.1016/j.exer.2011.02.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 01/19/2011] [Accepted: 02/04/2011] [Indexed: 12/30/2022]
|
21
|
Nickerson PEB, McLeod MC, Myers T, Clarke DB. Effects of epidermal growth factor and erythropoietin on Müller glial activation and phenotypic plasticity in the adult mammalian retina. J Neurosci Res 2011; 89:1018-30. [PMID: 21484851 DOI: 10.1002/jnr.22629] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Revised: 01/20/2011] [Accepted: 01/20/2011] [Indexed: 11/07/2022]
Abstract
Retinal Müller glia have received considerable attention with regard to their potential to function as quiescent retinal precursors. Various activation strategies induce characteristic features of retinal progenitor cells in Müller glia; however, these are often accompanied by hallmark features of reactive gliosis. We investigated the effects of an intravitreal injection of epidermal growth factor (EGF), a known mitogen, and erythropoietin (EPO) on activation and expression of developmental phenotypes within the adult retina. Using thymidine-analogue labeling as well as immunocytochemical and confocal analyses, we assayed the responses of retinal cells exposed to intravitreal administration of either EGF or EPO. We report that adult Müller glia incorporate bromodeoxyuridine (BrdU) and undergo a process of nuclear translocation to ectopic retinal layers following exposure to EGF. These cells survive within the retina for at least 23 days and express the developmental markers Pax6 and Chx10 as well as nestin and glial fibrillary acidic protein. Furthermore, we demonstrate that cotreatment with EGF and EPO suppresses aspects of EGF-induced glial reactivity, alters the retinal distribution of BrdU-positive nuclei, and serves to regulate the expression of developmental phenotypes seen in these cells. These data further our understanding of Müller cell responsiveness to intravitral, combinatorial growth factor treatments.
Collapse
Affiliation(s)
- P E B Nickerson
- Department of Biology, University of Victoria, Victoria, British Columbia, Canada
| | | | | | | |
Collapse
|
22
|
Phillips MJ, Otteson DC. Differential expression of neuronal genes in Müller glia in two- and three-dimensional cultures. Invest Ophthalmol Vis Sci 2011; 52:1439-49. [PMID: 21051699 DOI: 10.1167/iovs.10-6400] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Müller glia in the mammalian retina have some stem cell-like characteristics, although their capacity for neurogenesis remains limited both in vivo and in vitro. In vitro studies to date have used traditional two-dimensional (2D) cell culture to assess neuronal differentiation of Müller glia. The purpose of this study was to compare the effects of 2D and three-dimensional (3D) environments on Müller glial gene expression after growth factor stimulation. METHODS Conditionally immortalized mouse Müller glia cells (ImM10) were cultured under nonimmortalizing conditions with EGF/FGF2 to generate spheres that were differentiated in vitro on uncoated culture dishes (2D) or encapsulated in self-assembling, RADA-16 peptide hydrogels (3D) under identical media and growth factor supplementation conditions. Gene expression was analyzed using quantitative RT-PCR and immunocytochemistry. Cellular morphology was analyzed with light and confocal microscopy; sphere ultrastructure was analyzed with transmission electron microscopy. RESULTS ImM10 Müller cells express numerous genes associated with neural stem cells and retinal progenitors in both normal growth conditions and sphere-forming conditions. When encapsulated in the 3D hydrogel, cells can migrate and send processes into the hydrogel. Many genes associated with neurogenesis, as well as retinal neuron-specific genes, are differentially expressed in 2D and 3D differentiation conditions. CONCLUSIONS ImM10 Müller glia upregulate genes characteristic of retinal neurons after growth factor stimulation in vitro, and gene expression patterns are altered in 3D hydrogel cultures.
Collapse
Affiliation(s)
- M Joseph Phillips
- Department of Basic Sciences, College of Optometry, University of Houston, Houston, Texas 77204-2020, USA
| | | |
Collapse
|
23
|
Otteson DC, Phillips MJ. A conditional immortalized mouse muller glial cell line expressing glial and retinal stem cell genes. Invest Ophthalmol Vis Sci 2010; 51:5991-6000. [PMID: 20505190 DOI: 10.1167/iovs.10-5395] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
PURPOSE Müller glia have multiple functions in the retina, including synthesis of neurotrophic factors, uptake and metabolism of neurotransmitters, spatial buffering of ions, maintenance of the blood-retinal barrier, and response to injury. A population of Müller glia has some stem cell-like characteristics both in vivo and in vitro. The purpose of this study was to generate and characterize novel Müller glial cell lines from the postnatal mouse retina. METHODS Cells were cultured from postnatal day (P) 10 double heterozygous transgenic (H-2K(b)-tsA58/+; HRhoGFP/+) or C57BL/6 mice after papain dissociation. Interferon gamma (IFNγ) induction of the SV40 T-antigen (TAg) was assayed by immunohistochemistry and Western blot analysis. Proliferation was assayed by BrdU uptake and cell counts of calcein AM/ethidium bromide-stained cells. Gene expression was analyzed by RT-PCR and immunohistochemistry. RESULTS Conditionally immortalized (ImM10 [Immortmouse Müller P10]) and spontaneously immortalized (C57M10 [C57BL/6 Müller P10]) Müller glial cell lines were selected by differential adherence to laminin; both consisted of adherent flat cells with large, diffusely staining nuclei and an epithelial morphology. TAg induction stimulated BrdU uptake by Müller glia in mixed retinal cultures from H-2K(b)-tsA58/+; HRhoGFP/+ mice and increased the proliferation of ImM10 cells. ImM10 and C57M10 cells expressed genes characteristic of Müller glia but not genes characteristic of differentiated retinal neurons. ImM10 cells also expressed retinal stem cell genes. CONCLUSIONS The ImM10 cell line is a novel, conditionally immortalized Müller glial cell line isolated from the P10 mouse retina that expresses genes characteristic of Müller glial and retinal stem cells.
Collapse
Affiliation(s)
- Deborah C Otteson
- Department of Vision Science, College of Optometry, University of Houston, Houston, Texas 77204-2020, USA.
| | | |
Collapse
|
24
|
Moon SW, Chung EJ, Jung SA, Lee JH. PDGF stimulation of Müller cell proliferation: Contributions of c-JNK and the PI3K/Akt pathway. Biochem Biophys Res Commun 2009; 388:167-71. [PMID: 19653997 DOI: 10.1016/j.bbrc.2009.07.144] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Accepted: 07/28/2009] [Indexed: 12/15/2022]
Abstract
Platelet-derived growth factor (PDGF) has a critical role in proliferative vitreoretinopathy (PVR) as a chemoattractant and mitogen for retinal pigment epithelial cells and retinal glial cells. Here, we investigated the potential effects of PDGF on the proliferation of Müller cells and the intracellular signaling pathway mediating these changes. PDGF induced Müller cell proliferation and increased phosphorylation of the PDGF receptor (PDGFR), as shown by an MTT assay and immunoprecipitation analyses. Both effects were blocked by JNJ, a PDGFR-selective tyrosine kinase inhibitor. PDGF also stimulated phosphorylation of c-JNK and Akt. PDGF-induced Müller cell proliferation was significantly reduced by pre-treatment with SP600125 and LY294002, inhibitors of c-JNK and Akt phosphorylation, respectively. Our findings collectively indicate that PDGF-stimulated Müller cell proliferation occurs via activation of the c-JNK and PI3K/Akt signaling pathways. These data provide useful information in establishing the role of Müller cells in the development of proliferative vitreoretinopathy.
Collapse
Affiliation(s)
- Sang Woong Moon
- Department of Ophthalmology, Seoul Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | | | | | | |
Collapse
|
25
|
West E, Pearson R, MacLaren R, Sowden J, Ali R. Cell transplantation strategies for retinal repair. PROGRESS IN BRAIN RESEARCH 2009; 175:3-21. [PMID: 19660645 PMCID: PMC3272389 DOI: 10.1016/s0079-6123(09)17501-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell transplantation is a novel therapeutic strategy to restore visual responses to the degenerate adult neural retina and represents an exciting area of regenerative neurotherapy. So far, it has been shown that transplanted postmitotic photoreceptor precursors are able to functionally integrate into the adult mouse neural retina. In this review, we discuss the differentiation of photoreceptor cells from both adult and embryonic-derived stem cells and their potential for retinal cell transplantation. We also discuss the strategies used to overcome barriers present in the degenerate neural retina and improve retinal cell integration. Finally, we consider the future translation of retinal cell therapy as a therapeutic strategy to treat retinal degeneration.
Collapse
Affiliation(s)
- E.L. West
- Department of Genetics, UCL Institute of Ophthalmology, London, UK
| | - R.A. Pearson
- Department of Genetics, UCL Institute of Ophthalmology, London, UK
| | - R.E. MacLaren
- Department of Genetics, UCL Institute of Ophthalmology, London, UK
- Vitreoretinal Service, Moorfields Eye Hospital, London, UK
| | - J.C. Sowden
- Developmental Biology Unit, UCL Institute of Child Health, London, UK
| | - R.R. Ali
- Department of Genetics, UCL Institute of Ophthalmology, London, UK
- Molecular Immunology Unit, UCL Institute of Child Health, London, UK
| |
Collapse
|