Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(46): e2302089120, 2023 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-37931105

RESUMO

Ongoing cell therapy trials have demonstrated the need for precision control of donor cell behavior within the recipient tissue. We present a methodology to guide stem cell-derived and endogenously regenerated neurons by engineering the microenvironment. Being an "approachable part of the brain," the eye provides a unique opportunity to study neuron fate and function within the central nervous system. Here, we focused on retinal ganglion cells (RGCs)-the neurons in the retina are irreversibly lost in glaucoma and other optic neuropathies but can potentially be replaced through transplantation or reprogramming. One of the significant barriers to successful RGC integration into the existing mature retinal circuitry is cell migration toward their natural position in the retina. Our in silico analysis of the single-cell transcriptome of the developing human retina identified six receptor-ligand candidates, which were tested in functional in vitro assays for their ability to guide human stem cell-derived RGCs. We used our lead molecule, SDF1, to engineer an artificial gradient in the retina, which led to a 2.7-fold increase in donor RGC migration into the ganglion cell layer (GCL) and a 3.3-fold increase in the displacement of newborn RGCs out of the inner nuclear layer. Only donor RGCs that migrated into the GCL were found to express mature RGC markers, indicating the importance of proper structure integration. Together, these results describe an "in silico-in vitro-in vivo" framework for identifying, selecting, and applying soluble ligands to control donor cell function after transplantation.


Assuntos
Retina , Células Ganglionares da Retina , Recém-Nascido , Humanos , Células-Tronco , Neurogênese , Movimento Celular
2.
Mol Ther ; 32(5): 1425-1444, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38504518

RESUMO

Pathological ocular angiogenesis has long been associated with myeloid cell activation. However, the precise cellular and molecular mechanisms governing the intricate crosstalk between the immune system and vascular changes during ocular neovascularization formation remain elusive. In this study, we demonstrated that the absence of the suppressor of cytokine signaling 3 (SOCS3) in myeloid cells led to a substantial accumulation of microglia and macrophage subsets during the neovascularization process. Our single-cell RNA sequencing data analysis revealed a remarkable increase in the expression of the secreted phosphoprotein 1 (Spp1) gene within these microglia and macrophages, identifying subsets of Spp1-expressing microglia and macrophages during neovascularization formation in angiogenesis mouse models. Notably, the number of Spp1-expressing microglia and macrophages exhibited further elevation during neovascularization in mice lacking myeloid SOCS3. Moreover, our investigation unveiled the Spp1 gene as a direct transcriptional target gene of signal transducer and activator of transcription 3. Importantly, pharmaceutical activation of SOCS3 or blocking of SPP1 resulted in a significant reduction in pathological neovascularization. In conclusion, our study highlights the pivotal role of the SOCS3/STAT3/SPP1 axis in the regulation of pathological retinal angiogenesis.


Assuntos
Modelos Animais de Doenças , Macrófagos , Microglia , Osteopontina , Neovascularização Retiniana , Fator de Transcrição STAT3 , Proteína 3 Supressora da Sinalização de Citocinas , Animais , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/genética , Macrófagos/metabolismo , Camundongos , Microglia/metabolismo , Neovascularização Retiniana/metabolismo , Neovascularização Retiniana/patologia , Neovascularização Retiniana/genética , Neovascularização Retiniana/etiologia , Osteopontina/metabolismo , Osteopontina/genética , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT3/genética , Regulação da Expressão Gênica , Transdução de Sinais , Camundongos Knockout , Neovascularização Patológica/metabolismo , Neovascularização Patológica/genética , Angiogênese
3.
Angiogenesis ; 2024 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-38483712

RESUMO

Pathological retinal angiogenesis profoundly impacts visual function in vascular eye diseases, such as retinopathy of prematurity (ROP) in preterm infants and age-related macular degeneration in the elderly. While the involvement of photoreceptors in these diseases is recognized, the underlying mechanisms remain unclear. This study delved into the pivotal role of photoreceptors in regulating abnormal retinal blood vessel growth using an oxygen-induced retinopathy (OIR) mouse model through the c-Fos/A disintegrin and metalloprotease 17 (Adam17) axis. Our findings revealed a significant induction of c-Fos expression in rod photoreceptors, and c-Fos depletion in these cells inhibited pathological neovascularization and reduced blood vessel leakage in the OIR mouse model. Mechanistically, c-Fos directly regulated the transcription of Adam17 a shedding protease responsible for the production of bioactive molecules involved in inflammation, angiogenesis, and cell adhesion and migration. Furthermore, we demonstrated the therapeutic potential by using an adeno-associated virus carrying a rod photoreceptor-specific short hairpin RNA against c-fos which effectively mitigated abnormal retinal blood vessel overgrowth, restored retinal thickness, and improved electroretinographic (ERG) responses. In conclusion, this study highlights the significance of photoreceptor c-Fos in ROP pathology, offering a novel perspective for the treatment of this disease.

4.
J Cell Mol Med ; 26(11): 3254-3268, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35481949

RESUMO

Despite notable efforts and significant therapeutical advances, age-related macular degeneration remains the single most common reason for vision loss. Retinal progenitor cells (RPCs) are considered promising candidates for cellular treatments that repair and restore vision. In this allogenic study, the phenotypic profile of pig and human RPCs derived using similar manufacturing processes is compared. The long-term (12-week) survival of green fluorescent protein-pig retinal progenitor cells GFP-pRPC after subretinal transplantation into normal miniature pig (mini-pig) retina is investigated. Human eyes are both anatomically and physiologically mimicked by pig eyes, so the pig is an ideal model to show an equivalent way of delivering cells, immunological response and dosage. The phenotypic equivalency of porcine and clinically intended human RPCs was established. Thirty-nine mini-pigs are used in this study, and vehicle-injected eyes and non-injected eyes serve as controls. Six groups are given different dosages of pRPCs, and the cells are found to survive well in all groups. At 12 weeks, strong evidence of integration is indicated by the location of the grafted cells within the neuro-retina, extension of processes to the plexiform layers and expression of key retinal markers such as recoverin, rhodopsin and synaptophysin. No immunosuppression is used, and no immune response is found in any of the groups. No pRPC-related histopathology findings are reported in the major organs investigated. An initial dose of 250 k cells in 100 µl of buffer is established as an appropriate initial dose for future human clinical trials.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Retina , Animais , Diferenciação Celular/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Retina/metabolismo , Transplante de Células-Tronco , Suínos , Porco Miniatura
5.
Mol Vis ; 24: 733-745, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30581280

RESUMO

Purpose: To evaluate the potential of a poly(lactic-co-glycolic acid) (PLGA)-based slow release formulation of glial cell line-derived neurotrophic factor (GDNF) alone or in combination with melatonin to rescue photoreceptors in a mouse model of retinal degeneration. Methods: GDNF and GDNF/melatonin-loaded PLGA microspheres (MSs) were prepared using a solid-in-oil-in-water emulsion solvent extraction-evaporation technique. A combination of PLGA and vitamin E (VitE) was used to create the microcarriers. The structure, particle size, encapsulation efficiency, and in vitro release profile of the microparticulate formulations were characterized. Microparticulate systems (non-loaded, GDNF, and GDNF/melatonin-loaded MSs) were administered intravitreally to 3-week-old rhodopsin knockout mice (rho (-/-); n=7). The functional neuroprotective effect was assessed with electroretinography at 6, 9, and 12 weeks old. The rescue of the structure was determined with photoreceptor quantification at 12 weeks (9 weeks after administration of MSs). Immunohistochemistry for photoreceptor, glial, and proliferative markers was also performed. Results: The microspheres were able to deliver GDNF or to codeliver GDNF and melatonin in a sustained manner. Intravitreal injection of GDNF or GDNF/melatonin-loaded MSs led to partial functional and structural rescue of photoreceptors compared to blank microspheres or vehicle. No significant intraocular inflammatory reaction was observed after intravitreal injection of the microspheres. Conclusions: A single intravitreal injection of GDNF or GDNF/melatonin-loaded microspheres in the PLGA/VitE combination promoted the rescue of the photoreceptors in rho (-/-) mice. These intraocular drug delivery systems enable the efficient codelivery of therapeutically active substances for the treatment of retinal diseases.


Assuntos
Preparações de Ação Retardada/farmacocinética , Fator Neurotrófico Derivado de Linhagem de Célula Glial/farmacocinética , Melatonina/farmacocinética , Retina/efeitos dos fármacos , Degeneração Retiniana/terapia , Rodopsina/genética , Animais , Preparações de Ação Retardada/química , Modelos Animais de Doenças , Combinação de Medicamentos , Composição de Medicamentos/métodos , Liberação Controlada de Fármacos , Eletrorretinografia , Expressão Gênica , Injeções Intravítreas , Camundongos , Camundongos Knockout , Microesferas , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Retina/metabolismo , Retina/patologia , Degeneração Retiniana/genética , Degeneração Retiniana/metabolismo , Degeneração Retiniana/patologia , Rodopsina/agonistas , Rodopsina/deficiência , Vitamina E/química , Corpo Vítreo
6.
Adv Exp Med Biol ; 1074: 619-624, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29721995

RESUMO

Traumatic, inherited, and age-related degenerative diseases of the retina, such as retinal detachment, glaucoma, retinitis pigmentosa, and age-related macular degeneration, are characterized by the irreversible loss of retinal neurons. Several growth factors, including glial cell-derived neurotrophic factor and pigment epithelium-derived factor, have been shown to rescue retinal neurons in animal models of retinal disease. Here we describe a scalable and robust system to study the growth factor induction in the retina: retinal organoids derived from the induced pluripotent stem cells. We have demonstrated that they secrete GDNF and PEDF at the levels tenfold above detection limit for ELISA. We also have shown that growth factor production in this system may be upregulated by specific trigger, demonstrating the feasibility of this approach for drug discovery.


Assuntos
Proteínas do Olho/biossíntese , Células-Tronco Pluripotentes Induzidas , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Retina/metabolismo , Animais , Descoberta de Drogas/métodos , Proteínas do Olho/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/biossíntese , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Camundongos , Morfogênese , Fatores de Crescimento Neural/biossíntese , Fatores de Crescimento Neural/metabolismo , Organoides/metabolismo , Retina/citologia , Serpinas/biossíntese , Serpinas/metabolismo , Engenharia Tecidual/métodos
7.
J Biol Chem ; 289(10): 6362-6371, 2014 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-24407289

RESUMO

Cell transplantation is a potential therapeutic strategy for retinal degenerative diseases involving the loss of photoreceptors. However, it faces challenges to clinical translation due to safety concerns and a limited supply of cells. Human retinal progenitor cells (hRPCs) from fetal neural retina are expandable in vitro and maintain an undifferentiated state. This study aimed to investigate the therapeutic potential of hRPCs transplanted into a Royal College of Surgeons (RCS) rat model of retinal degeneration. At 12 weeks, optokinetic response showed that hRPC-grafted eyes had significantly superior visual acuity compared with vehicle-treated eyes. Histological evaluation of outer nuclear layer (ONL) characteristics such as ONL thickness, spread distance, and cell count demonstrated a significantly greater preservation of the ONL in hRPC-treated eyes compared with both vehicle-treated and control eyes. The transplanted hRPCs arrested visual decline over time in the RCS rat and rescued retinal morphology, demonstrating their potential as a therapy for retinal diseases. We suggest that the preservation of visual acuity was likely achieved through host photoreceptor rescue. We found that hRPC transplantation into the subretinal space of RCS rats was well tolerated, with no adverse effects such as tumor formation noted at 12 weeks after treatment.


Assuntos
Células-Tronco Embrionárias/transplante , Epitélio Pigmentado Ocular/transplante , Retina , Degeneração Retiniana/cirurgia , Transplante de Células-Tronco , Animais , Separação Celular , Células Cultivadas , Modelos Animais de Doenças , Feto/citologia , Humanos , Ratos , Retina/citologia , Retina/embriologia , Retina/fisiologia , Degeneração Retiniana/fisiopatologia , Acuidade Visual
8.
Mol Vis ; 20: 1271-80, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25352736

RESUMO

PURPOSE: The purpose of this study is to evaluate the functional and morphological changes in subretinal xenografts of human retinal progenitor cells (hRPCs) in B6 mice treated with Cyclosporin A (CsA; 210 mg/l in drinking water). METHODS: The hRPCs from human fetal eyes were isolated and expanded for transplantation. These cells, with green fluorescent protein (GFP) at 11 passages, were transplanted into the subretinal space in B6 mice. A combination of invasive and noninvasive approaches was used to analyze the structural and functional consequences of the subretinal injection of the hRPCs. The process of change was monitored using spectral domain optical coherence tomography (SDOCT), histology, and electroretinography (ERG) at 3 days, 1 week, and 3 weeks after transplantation. Cell counts were used to evaluate the survival rate with a confocal microscope. ERGs were performed to evaluate the physiologic changes, and the structural changes were evaluated using SDOCT and histological examination. RESULTS: The results of the histological examination showed that the hRPCs gained a better survival rate in the mice treated with CsA. The SDOCT showed that the bleb size of the retinal detachment was significantly decreased, and the retinal reattachment was nearly complete by 3 weeks. The ERG response amplitudes in the CsA group were less decreased after the injection, when compared with the control group, in the dark-adapted and light-adapted conditions. However, the cone-mediated function in both groups was less affected by the transplantation after 3 weeks than the rod-mediated function. CONCLUSIONS: Although significant functional and structural recovery was observed after the subretinal injection of the hRPCs, the effectiveness of CsA in xenotransplantation may be a novel and potential approach for increasing retinal progenitor cell survival.


Assuntos
Ciclosporina/farmacologia , Sobrevivência de Enxerto/efeitos dos fármacos , Imunossupressores/farmacologia , Retina/efeitos dos fármacos , Transplante de Células-Tronco , Células-Tronco/citologia , Animais , Proliferação de Células , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Eletrorretinografia , Feto , Genes Reporter , Sobrevivência de Enxerto/fisiologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Cultura Primária de Células , Retina/citologia , Retina/fisiologia , Retina/cirurgia , Células-Tronco/metabolismo , Tomografia de Coerência Óptica , Transplante Heterólogo
9.
bioRxiv ; 2024 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-38558999

RESUMO

Retinal ganglion cells (RGCs) lack regenerative capacity in mammals, and their degeneration in glaucoma leads to irreversible blindness. The transplantation of stem cell-derived RGCs lacks clinically relevant effect due to insufficient survival and integration of donor cells. We hypothesize that the retinal microenvironment plays a critical role in this process, and we can engineer a more acceptable setting for transplantation. Since the adult mammalian retina does not have regenerative capacity, we turned to the developing human retina to reconstruct cell-cell interactions at a single-cell level. We established a human fetal retina atlas by integrating currently available single-cell RNA sequencing datasets of human fetal retinas into a unified resource. We align RGC transcriptomes in pseudotime to map RGC developmental fate trajectories against the broader timeline of retinal development. Through this analysis, we identified brain-derived neurotrophic factor (BDNF) and glial-derived neurotrophic factor (GDNF) as key factors in RGC survival, highly expressed during fetal development but significantly reduced in adulthood despite the persistence of their receptors. To demonstrate the practical application of these findings, we show that using a slow-release formulation of BDNF and GDNF enhances RGC differentiation, survival, and function in vitro and improves RGC transplantation outcomes in a mouse model. BNDF/GDNF co-treatment not only increased survival and coverage of donor RGCs within the retina but also showed neuroprotective effects on host RGCs, preserving retinal function in a model of optic neuropathy. Altogether, our findings suggest that manipulating the retinal microenvironment with slow-release neurotrophic factors holds promise in regenerative medicine for treating glaucoma and other optic neuropathies. This approach not only improves donor cell survival and integration but also provides a neuroprotective benefit to host cells, indicating a significant advancement for glaucoma therapies.

10.
Acta Neuropathol Commun ; 12(1): 102, 2024 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-38907342

RESUMO

Neurofibromatosis Type 1 (NF1) is caused by loss of function variants in the NF1 gene. Most patients with NF1 develop skin lesions called cutaneous neurofibromas (cNFs). Currently the only approved therapeutic for NF1 is selumetinib, a mitogen -activated protein kinase (MEK) inhibitor. The purpose of this study was to analyze the transcriptome of cNF tumors before and on selumetinib treatment to understand both tumor composition and response. We obtained biopsy sets of tumors both pre- and on- selumetinib treatment from the same individuals and were able to collect sets from four separate individuals. We sequenced mRNA from 5844 nuclei and identified 30,442 genes in the untreated group and sequenced 5701 nuclei and identified 30,127 genes in the selumetinib treated group. We identified and quantified distinct populations of cells (Schwann cells, fibroblasts, pericytes, myeloid cells, melanocytes, keratinocytes, and two populations of endothelial cells). While we anticipated that cell proportions might change with treatment, we did not identify any one cell population that changed significantly, likely due to an inherent level of variability between tumors. We also evaluated differential gene expression based on drug treatment in each cell type. Ingenuity pathway analysis (IPA) was also used to identify pathways that differ on treatment. As anticipated, we identified a significant decrease in ERK/MAPK signaling in cells including Schwann cells but most specifically in myeloid cells. Interestingly, there is a significant decrease in opioid signaling in myeloid and endothelial cells; this downward trend is also observed in Schwann cells and fibroblasts. Cell communication was assessed by RNA velocity, Scriabin, and CellChat analyses which indicated that Schwann cells and fibroblasts have dramatically altered cell states defined by specific gene expression signatures following treatment (RNA velocity). There are dramatic changes in receptor-ligand pairs following treatment (Scriabin), and robust intercellular signaling between virtually all cell types associated with extracellular matrix (ECM) pathways (Collagen, Laminin, Fibronectin, and Nectin) is downregulated after treatment. These response specific gene signatures and interaction pathways could provide clues for understanding treatment outcomes or inform future therapies.


Assuntos
Benzimidazóis , Matriz Extracelular , Células de Schwann , Transdução de Sinais , Neoplasias Cutâneas , Humanos , Células de Schwann/efeitos dos fármacos , Células de Schwann/metabolismo , Células de Schwann/patologia , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia , Benzimidazóis/farmacologia , Matriz Extracelular/metabolismo , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/genética , Transdução de Sinais/efeitos dos fármacos , Neurofibroma/genética , Neurofibroma/tratamento farmacológico , Neurofibroma/metabolismo , Neurofibroma/patologia , Feminino , Masculino , RNA-Seq , Pessoa de Meia-Idade , Adulto , Neurofibromatose 1/genética , Neurofibromatose 1/tratamento farmacológico , Neurofibromatose 1/patologia , Inibidores de Proteínas Quinases/farmacologia , Transcriptoma/efeitos dos fármacos
11.
iScience ; 26(4): 106361, 2023 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-37009209

RESUMO

Neuronal repopulation achieved through transplantation or transdifferentiation from endogenous sources holds tremendous potential for restoring function in chronic neurodegenerative disease or acute injury. Key to the evaluation of neuronal engraftment is the definitive discrimination of new or donor neurons from preexisting cells within the host tissue. Recent work has identified mechanisms by which genetically encoded donor cell reporters can be transferred to host neurons through intercellular material transfer. In addition, labeling transplanted and endogenously transdifferentiated neurons through viral vector transduction can yield misexpression in host cells in some circumstances. These issues can confound the tracking and evaluation of repopulated neurons in regenerative experimental paradigms. Using the retina as an example, we discuss common reasons for artifactual labeling of endogenous host neurons with donor cell reporters and suggest strategies to prevent erroneous conclusions based on misidentification of cell origin.

12.
bioRxiv ; 2023 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-38014168

RESUMO

The limited regenerative potential of the optic nerve in adult mammals presents a major challenge for restoring vision after optic nerve trauma or disease. The mechanisms of this regenerative failure are not fully understood1,2. Here, through small-molecule and genetic screening for epigenetic modulators3, we identify DNA methyltransferase 3a (DNMT3a) as a potent inhibitor of axon regeneration in mouse and human retinal explants. Selective suppression of DNMT3a in retinal ganglion cells (RGCs) by gene targeting or delivery of shRNA leads to robust, full-length regeneration of RGC axons through the optic nerve and restoration of vision in adult mice after nerve crush injury. Genome-wide bisulfite and transcriptome profiling in combination with single nucleus RNA-sequencing of RGCs revealed selective DNA demethylation and reactivation of genetic programs supporting neuronal survival and axonal growth/regeneration by DNMT3a deficiency. This was accompanied by the suppression of gene networks associated with apoptosis and inflammation. Our results identify DNMT3a as the central orchestrator of an RGC-intrinsic mechanism that limits optic nerve regeneration. Suppressing DNMT3a expression in RGCs unlocks the epigenetic switch for optic nerve regeneration and presents a promising therapeutic avenue for effectively reversing vision loss resulted from optic nerve trauma or diseases.

13.
Cell Rep ; 42(8): 112889, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37527036

RESUMO

Microglia shift toward an inflammatory phenotype during aging that is thought to exacerbate age-related neurodegeneration. The molecular and cellular signals that resolve neuroinflammation post-injury are largely undefined. Here, we exploit systems genetics methods based on the extended BXD murine reference family and identify IGFBPL1 as an upstream cis-regulator of microglia-specific genes to switch off inflammation. IGFBPL1 is expressed by mouse and human microglia, and higher levels of its expression resolve lipopolysaccharide-induced neuroinflammation by resetting the transcriptome signature back to a homeostatic state via IGF1R signaling. Conversely, IGFBPL1 deficiency or selective deletion of IGF1R in microglia shifts these cells to an inflammatory landscape and induces early manifestation of brain tauopathy and retinal neurodegeneration. Therapeutic administration of IGFBPL1 drives pro-homeostatic microglia and prevents glaucomatous neurodegeneration and vision loss in mice. These results identify IGFBPL1 as a master driver of the counter-inflammatory microglial modulator that presents an endogenous resolution of neuroinflammation to prevent neurodegeneration in eye and brain.


Assuntos
Microglia , Tauopatias , Camundongos , Animais , Humanos , Microglia/metabolismo , Doenças Neuroinflamatórias , Tauopatias/metabolismo , Inflamação/metabolismo , Encéfalo/metabolismo , Homeostase , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Proteínas Supressoras de Tumor/metabolismo
14.
Ophthalmol Sci ; 3(4): 100390, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38025164

RESUMO

Purpose: The Retinal Ganglion Cell (RGC) Repopulation, Stem Cell Transplantation, and Optic Nerve Regeneration (RReSTORe) consortium was founded in 2021 to help address the numerous scientific and clinical obstacles that impede development of vision-restorative treatments for patients with optic neuropathies. The goals of the RReSTORe consortium are: (1) to define and prioritize the most critical challenges and questions related to RGC regeneration; (2) to brainstorm innovative tools and experimental approaches to meet these challenges; and (3) to foster opportunities for collaborative scientific research among diverse investigators. Design and Participants: The RReSTORe consortium currently includes > 220 members spanning all career stages worldwide and is directed by an organizing committee comprised of 15 leading scientists and physician-scientists of diverse backgrounds. Methods: Herein, we describe the structure and organization of the RReSTORe consortium, its activities to date, and the perceived impact that the consortium has had on the field based on a survey of participants. Results: In addition to helping propel the field of regenerative medicine as applied to optic neuropathies, the RReSTORe consortium serves as a framework for developing large collaborative groups aimed at tackling audacious goals that may be expanded beyond ophthalmology and vision science. Conclusions: The development of innovative interventions capable of restoring vision for patients suffering from optic neuropathy would be transformative for the ophthalmology field, and may set the stage for functional restoration in other central nervous system disorders. By coordinating large-scale, international collaborations among scientists with diverse and complementary expertise, we are confident that the RReSTORe consortium will help to accelerate the field toward clinical translation. Financial Disclosures: Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.

15.
Mol Neurodegener ; 18(1): 64, 2023 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-37735444

RESUMO

Retinal ganglion cell (RGC) death in glaucoma and other optic neuropathies results in irreversible vision loss due to the mammalian central nervous system's limited regenerative capacity. RGC repopulation is a promising therapeutic approach to reverse vision loss from optic neuropathies if the newly introduced neurons can reestablish functional retinal and thalamic circuits. In theory, RGCs might be repopulated through the transplantation of stem cell-derived neurons or via the induction of endogenous transdifferentiation. The RGC Repopulation, Stem Cell Transplantation, and Optic Nerve Regeneration (RReSTORe) Consortium was established to address the challenges associated with the therapeutic repair of the visual pathway in optic neuropathy. In 2022, the RReSTORe Consortium initiated ongoing international collaborative discussions to advance the RGC repopulation field and has identified five critical areas of focus: (1) RGC development and differentiation, (2) Transplantation methods and models, (3) RGC survival, maturation, and host interactions, (4) Inner retinal wiring, and (5) Eye-to-brain connectivity. Here, we discuss the most pertinent questions and challenges that exist on the path to clinical translation and suggest experimental directions to propel this work going forward. Using these five subtopic discussion groups (SDGs) as a framework, we suggest multidisciplinary approaches to restore the diseased visual pathway by leveraging groundbreaking insights from developmental neuroscience, stem cell biology, molecular biology, optical imaging, animal models of optic neuropathy, immunology & immunotolerance, neuropathology & neuroprotection, materials science & biomedical engineering, and regenerative neuroscience. While significant hurdles remain, the RReSTORe Consortium's efforts provide a comprehensive roadmap for advancing the RGC repopulation field and hold potential for transformative progress in restoring vision in patients suffering from optic neuropathies.


Assuntos
Doenças do Nervo Óptico , Células Ganglionares da Retina , Animais , Humanos , Retina , Encéfalo , Diferenciação Celular , Mamíferos
16.
Mol Ther Methods Clin Dev ; 21: 180-198, 2021 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-33816648

RESUMO

Optic neuropathies, including glaucoma, are a group of neurodegenerative diseases, characterized by the progressive loss of retinal ganglion cells (RGCs), leading to irreversible vision loss. While previous studies demonstrated the potential to replace RGCs with primary neurons from developing mouse retinas, their use is limited clinically. We demonstrate successful transplantation of mouse induced pluripotent stem cell (miPSC)/mouse embryonic stem cell (mESC)-derived RGCs into healthy and glaucomatous mouse retinas, at a success rate exceeding 65% and a donor cell survival window of up to 12 months. Transplanted Thy1-GFP+ RGCs were able to polarize within the host retina and formed axonal processes that followed host axons along the retinal surface and entered the optic nerve head. RNA sequencing of donor RGCs re-isolated from host retinas at 24 h and 1 week post-transplantation showed upregulation of cellular pathways mediating axonal outgrowth, extension, and guidance. Additionally, we provide evidence of subtype-specific diversity within miPSC-derived RGCs prior to transplantation.

17.
Transl Vis Sci Technol ; 9(10): 24, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-33024617

RESUMO

Purpose: Three-dimensional strategy for the differentiation of pluripotent stem cells to the retina has been widely used to study retinal development, although the cell production and drug discovery applications are limited by the throughput. Here we attempted to scale up the protocol using a semiautomated approach. Methods: For the experiments we used the Rx-GFP mouse embryonic stem cell (mES) reporter cell line, specific for early retinal development and human embryonic stem cell line Brn3b-tdTomato, specific for retinal ganglion cells. To increase the throughput, we implemented automated media exchange using Thermo WellWash Versa with Thermo RapidStack robot. To analyze the rate of retinal differentiation in mouse stem-cell derived organoids we imaged the plates at day 10 of differentiation using Life Technologies EVOS Fl Auto. The automated image analysis of fluorescent images was performed with custom Python OpenCV script. Results: The implementation of a semiautomated approach significantly reduced the operator time needed: 34 minutes versus two hours for 960 organoids over the course of 25 days without any change in differentiation pattern and quantity of retinal differentiation. Automated image analysis showed that Forskolin treatment starting from day 1 leads to a significant increase in retinal field induction efficiency. Conclusions: Semiautomated approach can be applied to retinal tissue differentiation to increase the throughput of the protocol. We demonstrated that automated image analysis can be used to evaluate differentiation efficiency, as well as for troubleshooting and to study factors affecting retinal differentiation. Translational Relevance: Using robotic approach reduces the risk of human error and allows to perform all cycle of cell production in enclosed conditions, which is critical for GMP cell manufacture.


Assuntos
Células-Tronco Pluripotentes , Retina , Animais , Diferenciação Celular , Linhagem Celular , Camundongos , Organoides
18.
Front Cell Neurosci ; 14: 171, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32719585

RESUMO

We have developed a deep learning-based computer algorithm to recognize and predict retinal differentiation in stem cell-derived organoids based on bright-field imaging. The three-dimensional "organoid" approach for the differentiation of pluripotent stem cells (PSC) into retinal and other neural tissues has become a major in vitro strategy to recapitulate development. We decided to develop a universal, robust, and non-invasive method to assess retinal differentiation that would not require chemical probes or reporter gene expression. We hypothesized that basic-contrast bright-field (BF) images contain sufficient information on tissue specification, and it is possible to extract this data using convolutional neural networks (CNNs). Retina-specific Rx-green fluorescent protein mouse embryonic reporter stem cells have been used for all of the differentiation experiments in this work. The BF images of organoids have been taken on day 5 and fluorescent on day 9. To train the CNN, we utilized a transfer learning approach: ImageNet pre-trained ResNet50v2, VGG19, Xception, and DenseNet121 CNNs had been trained on labeled BF images of the organoids, divided into two categories (retina and non-retina), based on the fluorescent reporter gene expression. The best-performing classifier with ResNet50v2 architecture showed a receiver operating characteristic-area under the curve score of 0.91 on a test dataset. A comparison of the best-performing CNN with the human-based classifier showed that the CNN algorithm performs better than the expert in predicting organoid fate (84% vs. 67 ± 6% of correct predictions, respectively), confirming our original hypothesis. Overall, we have demonstrated that the computer algorithm can successfully recognize and predict retinal differentiation in organoids before the onset of reporter gene expression. This is the first demonstration of CNN's ability to classify stem cell-derived tissue in vitro.

19.
Cell Transplant ; 29: 963689720964383, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33356508

RESUMO

The optic pathway glioma (OPG) is a slow-growing brain tumor that arises along the optic nerve or its downstream connections and causing vision to gradually worsen with time. This tumor forms in children with a genetic condition called neurofibromatosis type 1 (NF1), causing tumors to grow on nerves. In normal conditions, glial cells are there to support and protect nerve cells but, in NF1-OPG, glial cells have a genetic defect and grow out of control forming a tumor called a glioma. There are no rat models of NF1-OPG that can be used to explore various treatment options, and mouse models make interventional studies difficult due to their small eye size. We have created a model in which to study the progression of tumor growth in the optic nerve and establish the anatomical and functional consequences of the model and determine its suitability to serve as a surrogate for human disease. C6 rat glioma cells were injected into the optic nerve of Long-Evans rats and allowed to proliferate for 2 weeks. The eye clearly showed proptosis and lens opacity was observed, likely due to increased intraocular pressure caused by growing tumors. Hematoxylin-eosin staining showed marked cellularity, with hyperchromatism and pleomorphism. There was prominent area of necrosis with neoplastic cells palisading around the penumbra. Immunostaining with markers such as S100, ß-tubulin III, Foxp3, CD45, Vimentin, and Ki67 confirmed low-grade tumor formation, with a mild immune response. Our results show the utility of a surgically induced rat model of OPG that may be used for exploring various treatment options for NF1 ocular tumors.


Assuntos
Glioma/metabolismo , Nervo Óptico/metabolismo , Doenças Retinianas/metabolismo , Linhagem Celular Tumoral , Citometria de Fluxo , Fatores de Transcrição Forkhead/metabolismo , Glioma/genética , Humanos , Antígeno Ki-67/genética , Antígeno Ki-67/metabolismo , Neurofibromina 1/metabolismo , Nervo Óptico/patologia , Doenças Retinianas/genética , Doenças Retinianas/patologia , Tubulina (Proteína)/metabolismo , Vimentina/metabolismo
20.
Tissue Eng Part C Methods ; 25(7): 433-445, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31195897

RESUMO

IMPACT STATEMENT: The development of retinal regenerative therapies relies on the reproducible and renewable source of retinal neurons for drug discovery and cell transplantation. Three-dimensional approach for retinal differentiation from pluripotent cells recently emerged as the robust strategy for retinal tissue differentiation. In this work, we present the combination of optimized conditions and techniques for three-dimensional retinal differentiation from mouse embryonic cells that improves reproducibility and efficiency of retinal differentiation in organoid cultures. We also show that the retinal induction can be achieved with the synthetic oligopeptide instead of Matrigel that allows to approach xeno-free conditions for cell production.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células-Tronco Pluripotentes/citologia , Retina/citologia , Animais , Agregação Celular , Linhagem Celular , Colágeno/farmacologia , Combinação de Medicamentos , Laminina/farmacologia , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Proteoglicanas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA