Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
2.
J Hematol Oncol ; 13(1): 62, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32487125

ABSTRACT

BACKGROUND: Expression of Oct4 maintains cancer stem cell (CSC)-like properties in lung cancer cells and is correlated with poor prognosis of lung adenocarcinoma. M2-type tumor-associated macrophages (TAMs) promote cancer cell migration and metastasis. Tumor microenvironments promote monocyte differentiation into M2 TAMs via a complex cytokine-based connection. We explored the role of Oct4 in cytokine secretion in lung cancer and its impact on M2 TAM polarization. METHODS: Monocytes co-cultured with the conditioned medium from Oct4-overexpressing lung cancer cells were used to investigate M2 TAM differentiation. The inflammatory factors in the conditioned medium of Oct4-overexpressing A549 cells were examined using human inflammation antibody arrays. The correlations of Oct4, macrophage colony-stimulating factor (M-CSF), and M2 TAMs were validated in lung cancer cells, syngeneic mouse lung tumor models, and clinical samples of non-small cell lung cancer (NSCLC). RESULTS: Oct4-overexpressing A549 cells expressed elevated levels of M-CSF, which contributed to increased M2 macrophages and enhanced tumor migration. Overexpression of Oct4 enhanced tumor growth and reduced the survival of lung tumor-bearing mice, which was correlated with increased number of M2 macrophages in lung cancer. Notably, NSCLC patients with high expression levels of Oct4, M-CSF, and M2 TAMs had the poorest recurrence-free survival. A positive correlation between Oct4, M-CSF, and M2 TAMs was observed in the tumor tissue of NSCLC patient. Treatment with all-trans retinoic acid exerted anti-tumor effects and reduced M2 TAMs in tumor-bearing mice. CONCLUSIONS: Our results indicate that Oct4 expressed by lung cancer cells promotes M2 macrophage polarization through upregulation of M-CSF secretion, leading to cancer growth and metastasis. Our findings also implicate that the Oct4/M-CSF axis in M2 macrophage polarization may be potential therapeutic targets for lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Macrophage Colony-Stimulating Factor/biosynthesis , Neoplasm Proteins/physiology , Octamer Transcription Factor-3/physiology , Tumor-Associated Macrophages/pathology , A549 Cells , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/mortality , Cell Differentiation , Cohort Studies , Culture Media, Conditioned/pharmacology , Cytokines/physiology , Genes, Reporter , Humans , Lung Neoplasms/mortality , Macrophage Colony-Stimulating Factor/genetics , Male , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Octamer Transcription Factor-3/antagonists & inhibitors , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/pharmacology , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Recombinant Proteins/pharmacology , THP-1 Cells , Tretinoin/pharmacology , Tumor Microenvironment , Up-Regulation/drug effects
3.
J Dent Res ; 97(1): 77-83, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28898112

ABSTRACT

The induced pluripotent stem cells (iPSCs) have an intrinsic capability for indefinite self-renewal and large-scale expansion and can differentiate into all types of cells. Here, we tested the potential of iPSCs from dental pulp stem cells (DPSCs) to differentiate into functional odontoblasts. DPSCs were reprogrammed into iPSCs via electroporation of reprogramming factors OCT-4, SOX2, KLF4, LIN28, and L-MYC. The iPSCs presented overexpression of the reprogramming genes and high protein expressions of alkaline phosphatase, OCT4, and TRA-1-60 in vitro and generated tissues from 3 germ layers in vivo. Dentin discs with poly-L-lactic acid scaffolds containing iPSCs were implanted subcutaneously into immunodeficient mice. After 28 d from implantation, the iPSCs generated a pulp-like tissue with the presence of tubular dentin in vivo. The differentiation potential after long-term expansion was assessed in vitro. iPSCs and DPSCs of passages 4 and 14 were treated with either odontogenic medium or extract of bioactive cement for 28 d. Regardless of the passage tested, iPSCs expressed putative markers of odontoblastic differentiation and kept the same mineralization potential, while DPSC P14 failed to do the same. Analysis of these data collectively demonstrates that human iPSCs can be a source to derive human odontoblasts for dental pulp research and test bioactivity of materials.


Subject(s)
Induced Pluripotent Stem Cells/physiology , Odontoblasts/physiology , Animals , Cell Differentiation/drug effects , Dental Pulp/drug effects , Dental Pulp/physiology , Electroporation , Female , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/pharmacology , Mice , Mice, SCID , Octamer Transcription Factor-3/pharmacology , Odontoblasts/metabolism , Proto-Oncogene Proteins c-myc/pharmacology , RNA-Binding Proteins/pharmacology , SOXB1 Transcription Factors/pharmacology , Tissue Scaffolds
4.
Stem Cell Res ; 15(1): 240-2, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26141785

ABSTRACT

The combination of OCT4 expression and short-term exposure to reprogramming media induces a state of transcriptional plasticity in human fibroblasts, capable of responding to changes in the extracellular environment. Here we provide characterization of iPSCs established through continued culture of OCT4-induced plastic human fibroblasts in pluripotent-supportive reprogramming media. Human iPSC(OCT4) are morphologically indistinguishable from conventionally derived iPSCs and express core proteins involved in maintenance of pluripotency. iPSC(OCT4) display bona fide functional pluripotency as measured by in vivo teratoma formation consisting of the three germ layers.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Octamer Transcription Factor-3/pharmacology , Adult , Animals , Cellular Reprogramming/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Induced Pluripotent Stem Cells/drug effects , Mice, Inbred NOD , Mice, SCID
5.
Stem Cell Res ; 15(1): 221-30, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26117529

ABSTRACT

The combination of OCT4 expression and short-term exposure to reprogramming media induces a state of transcriptional plasticity in human fibroblasts, capable of responding to changes in the extracellular environment that facilitate direct cell fate conversion toward lineage specific progenitors. Here we reveal that continued exposure of OCT4-induced plastic human fibroblasts to reprogramming media (RM) is sufficient to induce pluripotency. OCT4-derived induced pluripotent stem cell (iPSC(OCT4)) colonies emerged after prolonged culture in RM, and formed independently of lineage specific progenitors. Human iPSC(OCT4) are morphologically indistinguishable from conventionally derived iPSCs and express core proteins involved in maintenance of pluripotency. iPSC(OCT4) display in vivo functional pluripotency as measured by teratoma formation consisting of the three germ layers, and are capable of targeted in vitro differentiation. Our study indicates that acquisition of pluripotency is one of multiple cell fate choices that can be facilitated through environmental stimulation of OCT4-induced plasticity, and suggests the role of other reprogramming factors to induce pluripotency can be substituted by prolonged culture of plastic fibroblasts.


Subject(s)
Fibroblasts/cytology , Induced Pluripotent Stem Cells/cytology , Octamer Transcription Factor-3/pharmacology , Adult , Animals , Cell Lineage/drug effects , Fibroblasts/drug effects , Humans , Immunophenotyping , Mice, Inbred NOD , Mice, SCID , Models, Biological
6.
PLoS One ; 9(7): e102510, 2014.
Article in English | MEDLINE | ID: mdl-25019378

ABSTRACT

From the cocktail of four factors that were able to induce pluripotent stem cells from differentiated cells, Oct-4, c-Myc, Sox-2 and Klf4, only Oct-4 was not expressed during regeneration in newts. To explore the possible action of this stemness factor we developed an assay where we introduced exogenous Oct-4 protein to an in vitro system for lens regeneration in newts. We found that exogenous Oct-4 inhibits differentiation of iris pigmented epithelial cells into lens cells and also regulates Sox-2 and Pax-6, both important players during lens development. Thus, presence of Oct-4 hinders transdifferentiation of iris cells.


Subject(s)
Cell Transdifferentiation/drug effects , Lens, Crystalline/drug effects , Octamer Transcription Factor-3/pharmacology , Salamandridae/growth & development , Animals , Eye Proteins/genetics , Eye Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lens, Crystalline/cytology , PAX6 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , Recombinant Proteins/pharmacology , Regeneration/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Salamandridae/genetics , Salamandridae/physiology
7.
Eur Heart J ; 34(21): 1575-86, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22621821

ABSTRACT

AIMS: Myocardial cell replacement therapies are hampered by a paucity of sources for human cardiomyocytes and by the expected immune rejection of allogeneic cell grafts. The ability to derive patient-specific human-induced pluripotent stem cells (hiPSCs) may provide a solution to these challenges. We aimed to derive hiPSCs from heart failure (HF) patients, to induce their cardiomyocyte differentiation, to characterize the generated hiPSC-derived cardiomyocytes (hiPSC-CMs), and to evaluate their ability to integrate with pre-existing cardiac tissue. METHODS AND RESULTS: Dermal fibroblasts from two HF patients were reprogrammed by retroviral delivery of Oct4, Sox2, and Klf4 or by using an excisable polycistronic lentiviral vector. The resulting HF-hiPSCs displayed adequate reprogramming properties and could be induced to differentiate into cardiomyocytes with the same efficiency as control hiPSCs (derived from human foreskin fibroblasts). Gene expression and immunostaining studies confirmed the cardiomyocyte phenotype of the differentiating HF-hiPSC-CMs. Multi-electrode array recordings revealed the development of a functional cardiac syncytium and adequate chronotropic responses to adrenergic and cholinergic stimulation. Next, functional integration and synchronized electrical activities were demonstrated between hiPSC-CMs and neonatal rat cardiomyocytes in co-culture studies. Finally, in vivo transplantation studies in the rat heart revealed the ability of the HF-hiPSC-CMs to engraft, survive, and structurally integrate with host cardiomyocytes. CONCLUSIONS: Human-induced pluripotent stem cells can be established from patients with advanced heart failure and coaxed to differentiate into cardiomyocytes, which can integrate with host cardiac tissue. This novel source for patient-specific heart cells may bring a unique value to the emerging field of cardiac regenerative medicine.


Subject(s)
Heart Failure/pathology , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Animals , Cell Differentiation , Cell Survival , Cellular Reprogramming/drug effects , Female , Genetic Vectors , Heart Failure/therapy , Humans , Induced Pluripotent Stem Cells/transplantation , Karyotype , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/pharmacology , Octamer Transcription Factor-3/pharmacology , Rats , Rats, Sprague-Dawley , SOXB1 Transcription Factors/pharmacology , Transgenes , Transplantation, Heterologous
8.
Stem Cells ; 31(2): 259-68, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23136034

ABSTRACT

MicroRNAs (miRNAs) have emerged as critical regulators of gene expression through translational inhibition and RNA decay and have been implicated in the regulation of cellular differentiation, proliferation, angiogenesis, and apoptosis. In this study, we analyzed global miRNA and mRNA microarrays to predict novel miRNA-mRNA interactions in human embryonic stem cells and induced pluripotent stem cells (iPSCs). In particular, we demonstrate a regulatory feedback loop between the miR-302 cluster and two transcription factors, NR2F2 and OCT4. Our data show high expression of miR-302 and OCT4 in pluripotent cells, while NR2F2 is expressed exclusively in differentiated cells. Target analysis predicts that NR2F2 is a direct target of miR-302, which we experimentally confirm by reporter luciferase assays and real-time polymerase chain reaction. We also demonstrate that NR2F2 directly inhibits the activity of the OCT4 promoter and thus diminishes the positive feedback loop between OCT4 and miR-302. Importantly, higher reprogramming efficiencies were obtained when we reprogrammed human adipose-derived stem cells into iPSCs using four factors (KLF4, C-MYC, OCT4, and SOX2) plus miR-302 (this reprogramming cocktail is hereafter referred to as "KMOS3") when compared to using four factors ("KMOS"). Furthermore, shRNA knockdown of NR2F2 mimics the over-expression of miR-302 by also enhancing reprogramming efficiency. Interestingly, we were unable to generate iPSCs from miR-302a/b/c/d alone, which is in contrast to previous publications that have reported that miR-302 by itself can reprogram human skin cancer cells and human hair follicle cells. Taken together, these findings demonstrate that miR-302 inhibits NR2F2 and promotes pluripotency through indirect positive regulation of OCT4. This feedback loop represents an important new mechanism for understanding and inducing pluripotency in somatic cells.


Subject(s)
Adipocytes/drug effects , COUP Transcription Factor II/genetics , Induced Pluripotent Stem Cells/drug effects , MicroRNAs/genetics , Octamer Transcription Factor-3/genetics , Adipocytes/cytology , Adipocytes/metabolism , COUP Transcription Factor II/antagonists & inhibitors , COUP Transcription Factor II/metabolism , Cell Differentiation/drug effects , Cellular Reprogramming/drug effects , Cellular Reprogramming/genetics , Feedback, Physiological , Female , Gene Expression Regulation/drug effects , Genes, Reporter , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Kruppel-Like Transcription Factors/pharmacology , Luciferases , MicroRNAs/metabolism , MicroRNAs/pharmacology , Microarray Analysis , Octamer Transcription Factor-3/metabolism , Octamer Transcription Factor-3/pharmacology , Primary Cell Culture , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/pharmacology , RNA, Small Interfering/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/pharmacology , Signal Transduction/drug effects
9.
Cell Biochem Biophys ; 66(2): 309-18, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23212180

ABSTRACT

Fibroblasts can be reprogrammed by ectopic expression of reprogramming factors to yield induced pluripotent stem (iPS) cells that are capable of transdifferentiating into diverse types of somatic cell lines. In this study, we examined if functional cardiomyocytes (CMs) can be produced from mouse cardiac fibroblasts (CFs), using iPS cell factor-based reprogramming. CFs were isolated from Oct4-GFP-C57 mice and infected with a retrovirus expressing the Yamanaka reprogramming factors, Oct4, Sox2, Klf4, and c-Myc to reprogram the CFs into a CF-iPS cell line. Primary mouse embryonic fibroblast cells (MEFs) were used as a control. We found that the dedifferentiated CF-iPS cells showed similar biological characteristics (morphology, pluripotent factor expression, and methylation level) as embryonic stem cells (ESs) and MEF-iPS cells. We used the classical embryoid bodies (EBs)-based method and a transwell CM co-culture system to simulate the myocardial paracrine microenvironment for performing CF-iPS cell cardiogenic differentiation. Under this simulated myocardial microenvironment, CF-iPS cells formed spontaneously beating EBs. The transdifferentiated self-beating cells expressed cardiac-specific transcription and structural factors and also displayed typical myocardial morphology and electrophysiological characteristics. CFs can be dedifferentiated into iPS cells and further transdifferentiated into CMs. CFs hold great promise for CM regeneration as an autologous cell source for functional CM in situ without the need for exogenous cell transplantation in ischemic heart disease.


Subject(s)
Cellular Reprogramming , Fibroblasts/cytology , Myocytes, Cardiac/cytology , Animals , Cell Differentiation , Cells, Cultured , Cellular Reprogramming/drug effects , Coculture Techniques , Embryoid Bodies/physiology , Fibroblasts/drug effects , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/pharmacology , Mice , Mice, Inbred C57BL , Octamer Transcription Factor-3/pharmacology , Proto-Oncogene Proteins c-myc/pharmacology , SOXB1 Transcription Factors/pharmacology
10.
Mol Vis ; 18: 2871-81, 2012.
Article in English | MEDLINE | ID: mdl-23233789

ABSTRACT

PURPOSE: This study aimed to develop a feasible and efficient method for generating embryonic stem cell (ESC)-like induced pluripotent stem (iPS) cells from human Tenon's capsule fibroblasts (HTFs) through the expression of a defined set of transcription factors, which will have significant application value for ophthalmic personalized regenerative medicine. METHODS: HTFs were harvested from fresh samples, and reprogramming was induced by the exogenous expression of the four classic transcription factors, OCT-3/4, SOX-2, KLF-4, and C-MYC. The HTF-derived iPS (TiPS) cells were analyzed with phase contrast microscopy, real-time PCR, immunofluorescence, FACS analysis, alkaline phosphatase activity analysis, and a teratoma formation assay. Human ESC colonies were used as the positive control. RESULTS: The resulting HTF-derived iPS cell colonies were indistinguishable from human ESC colonies regarding morphology, gene expression levels, pluripotent gene expression, alkaline phosphatase activity, and the ability to generate all three embryonic germ layers. CONCLUSIONS: This study presents a simple, efficient, practical procedure for generating patient-tailored iPS cells from HTFs. These cells will serve as a valuable and preferred candidate donor cell population for ophthalmological regenerative medicine.


Subject(s)
Cell Differentiation/drug effects , Fibroblasts/cytology , Induced Pluripotent Stem Cells/cytology , Tenon Capsule/cytology , Adolescent , Adult , Alkaline Phosphatase/metabolism , Biomarkers/metabolism , Cell Proliferation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Flow Cytometry , Germ Layers/cytology , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/pharmacology , Microscopy, Phase-Contrast , Middle Aged , Octamer Transcription Factor-3/pharmacology , Primary Cell Culture , Proto-Oncogene Proteins c-myc/pharmacology , Real-Time Polymerase Chain Reaction , SOXB1 Transcription Factors/pharmacology , Tenon Capsule/drug effects , Tenon Capsule/metabolism
11.
PLoS One ; 6(1): e16182, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-21249204

ABSTRACT

Epidermal melanocytes play an important role in protecting the skin from UV rays, and their functional impairment results in pigment disorders. Additionally, melanomas are considered to arise from mutations that accumulate in melanocyte stem cells. The mechanisms underlying melanocyte differentiation and the defining characteristics of melanocyte stem cells in humans are, however, largely unknown. In the present study, we set out to generate melanocytes from human iPS cells in vitro, leading to a preliminary investigation of the mechanisms of human melanocyte differentiation. We generated iPS cell lines from human dermal fibroblasts using the Yamanaka factors (SOX2, OCT3/4, and KLF4, with or without c-MYC). These iPS cell lines were subsequently used to form embryoid bodies (EBs) and then differentiated into melanocytes via culture supplementation with Wnt3a, SCF, and ET-3. Seven weeks after inducing differentiation, pigmented cells expressing melanocyte markers such as MITF, tyrosinase, SILV, and TYRP1, were detected. Melanosomes were identified in these pigmented cells by electron microscopy, and global gene expression profiling of the pigmented cells showed a high similarity to that of human primary foreskin-derived melanocytes, suggesting the successful generation of melanocytes from iPS cells. This in vitro differentiation system should prove useful for understanding human melanocyte biology and revealing the mechanism of various pigment cell disorders, including melanoma.


Subject(s)
Cell Differentiation/drug effects , DNA-Binding Proteins/pharmacology , Induced Pluripotent Stem Cells/cytology , Melanocytes/cytology , Cell Culture Techniques/methods , Embryoid Bodies , Fibroblasts/cytology , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/pharmacology , Octamer Transcription Factor-3/pharmacology , SOXB1 Transcription Factors/pharmacology , Skin/cytology
12.
Stem Cells Dev ; 19(5): 615-20, 2010 May.
Article in English | MEDLINE | ID: mdl-19527182

ABSTRACT

Reprogramming of mouse and human somatic cells into induced pluripotent stem (iPS) cells has been made possible with the expression of the transcription factor quartet Oct4, Sox2, c-Myc, and Klf4. Here, we compared iPS cells derived from mouse embryonic fibroblasts with the 4 factors to embryonic stem cells by electron microscopy. Both cell types are almost indistinguishable at the ultrastructural level, providing further evidence for the similarity of these 2 pluripotent stem cell populations.


Subject(s)
Cellular Reprogramming/drug effects , Induced Pluripotent Stem Cells/cytology , Transcription Factors/pharmacology , Animals , Cells, Cultured , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/pharmacology , Mice , Microscopy, Electron , Octamer Transcription Factor-3/pharmacology , Proto-Oncogene Proteins c-myc/pharmacology , SOXB1 Transcription Factors/pharmacology
13.
Stem Cells ; 27(10): 2427-34, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19658190

ABSTRACT

Human induced pluripotent stem cells (iPSCs) have great promise for cellular therapy, but it is unclear if they have the same potential as human embryonic stem cells (hESCs) to differentiate into specialized cell types. Ocular cells such as the retinal pigmented epithelium (RPE) are of particular interest because they could be used to treat degenerative eye diseases, including age-related macular degeneration and retinitis pigmentosa. We show here that iPSCs generated using Oct4, Sox2, Nanog, and Lin28 can spontaneously differentiate into RPE cells, which can then be isolated and cultured to form highly differentiated RPE monolayers. RPE derived from iPSCs (iPS-RPE) were analyzed with respect to gene expression, protein expression, and rod outer segment phagocytosis, and compared with cultured fetal human RPE (fRPE) and RPE derived from hESCs (hESC-RPE). iPS-RPE expression of marker mRNAs was quantitatively similar to that of fRPE and hESC-RPE, and marker proteins were appropriately expressed and localized in polarized monolayers. Levels of rod outer segment phagocytosis by iPS-RPE, fRPE, and hESC-RPE were likewise similar and dependent on integrin alpha v beta 5. This work shows that iPSCs can differentiate into functional RPE that are quantitatively similar to fRPE and hESC-RPE and further supports the finding that iPSCs are similar to hESCs in their differentiation potential.


Subject(s)
Cell Differentiation/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Retinal Pigment Epithelium/cytology , Retinal Pigment Epithelium/metabolism , Biomarkers/analysis , Biomarkers/metabolism , Brain Tissue Transplantation/methods , Cell Differentiation/drug effects , Cell Line , Cell Polarity/physiology , Cell Proliferation , Homeodomain Proteins/metabolism , Homeodomain Proteins/pharmacology , Humans , Integrin alphaV/metabolism , Nanog Homeobox Protein , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Octamer Transcription Factor-3/metabolism , Octamer Transcription Factor-3/pharmacology , Phagocytosis/physiology , Phenotype , Pluripotent Stem Cells/drug effects , RNA, Messenger/analysis , RNA, Messenger/metabolism , Regeneration/drug effects , Regeneration/physiology , Retinal Diseases/therapy , Retinal Rod Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/metabolism , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...