Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
Proc Natl Acad Sci U S A ; 120(42): e2308204120, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37812728

ABSTRACT

Migration is essential for the laminar stratification and connectivity of neurons in the central nervous system. In the retina, photoreceptors (PRs) migrate to positions according to birthdate, with early-born cells localizing to the basal-most side of the outer nuclear layer. It was proposed that apical progenitor mitoses physically drive these basal translocations non-cell autonomously, but direct evidence is lacking, and whether other mechanisms participate is unknown. Here, combining loss- or gain-of-function assays to manipulate cell cycle regulators (Sonic hedgehog, Cdkn1a/p21) with an in vivo lentiviral labelling strategy, we demonstrate that progenitor division is one of two forces driving basal translocation of rod soma. Indeed, replacing Shh activity rescues abnormal rod translocation in retinal explants. Unexpectedly, we show that rod differentiation also promotes rod soma translocation. While outer segment function or formation is dispensable, Crx and SNARE-dependent synaptic function are essential. Thus, both non-cell and cell autonomous mechanisms underpin PR soma sublaminar positioning in the mammalian retina.


Subject(s)
Neurosecretion , Retinal Rod Photoreceptor Cells , Animals , Retinal Rod Photoreceptor Cells/metabolism , Hedgehog Proteins/metabolism , Retina/metabolism , Cell Differentiation , Mammals
2.
EMBO J ; 41(4): e106825, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35023164

ABSTRACT

Despite extensive analysis of pRB phosphorylation in vitro, how this modification influences development and homeostasis in vivo is unclear. Here, we show that homozygous Rb∆K4 and Rb∆K7 knock-in mice, in which either four or all seven phosphorylation sites in the C-terminal region of pRb, respectively, have been abolished by Ser/Thr-to-Ala substitutions, undergo normal embryogenesis and early development, notwithstanding suppressed phosphorylation of additional upstream sites. Whereas Rb∆K4 mice exhibit telomere attrition but no other abnormalities, Rb∆K7 mice are smaller and display additional hallmarks of premature aging including infertility, kyphosis, and diabetes, indicating an accumulative effect of blocking pRb phosphorylation. Diabetes in Rb∆K7 mice is insulin-sensitive and associated with failure of quiescent pancreatic ß-cells to re-enter the cell cycle in response to mitogens, resulting in induction of DNA damage response (DDR), senescence-associated secretory phenotype (SASP), and reduced pancreatic islet mass and circulating insulin level. Pre-treatment with the epigenetic regulator vitamin C reduces DDR, increases cell cycle re-entry, improves islet morphology, and attenuates diabetes. These results have direct implications for cell cycle regulation, CDK-inhibitor therapeutics, diabetes, and longevity.


Subject(s)
Aging/physiology , Ascorbic Acid/pharmacology , Diabetes Mellitus, Experimental/prevention & control , Retinoblastoma Protein/metabolism , Animals , Cellular Senescence/drug effects , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , E2F1 Transcription Factor/metabolism , Embryonic Development/genetics , Female , Fibroblasts/drug effects , Gene Knock-In Techniques , Insulin-Secreting Cells/pathology , Mice , Phosphorylation , Pregnancy , Retinoblastoma Protein/genetics , Telomere/genetics
3.
Cancer Cell ; 39(8): 1115-1134.e12, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34270926

ABSTRACT

Cancer heterogeneity impacts therapeutic response, driving efforts to discover over-arching rules that supersede variability. Here, we define pan-cancer binary classes based on distinct expression of YAP and YAP-responsive adhesion regulators. Combining informatics with in vivo and in vitro gain- and loss-of-function studies across multiple murine and human tumor types, we show that opposite pro- or anti-cancer YAP activity functionally defines binary YAPon or YAPoff cancer classes that express or silence YAP, respectively. YAPoff solid cancers are neural/neuroendocrine and frequently RB1-/-, such as retinoblastoma, small cell lung cancer, and neuroendocrine prostate cancer. YAP silencing is intrinsic to the cell of origin, or acquired with lineage switching and drug resistance. The binary cancer groups exhibit distinct YAP-dependent adhesive behavior and pharmaceutical vulnerabilities, underscoring clinical relevance. Mechanistically, distinct YAP/TEAD enhancers in YAPoff or YAPon cancers deploy anti-cancer integrin or pro-cancer proliferative programs, respectively. YAP is thus pivotal across cancer, but in opposite ways, with therapeutic implications.


Subject(s)
Lung Neoplasms/genetics , Small Cell Lung Carcinoma/genetics , TEA Domain Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/genetics , YAP-Signaling Proteins/genetics , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Humans , Integrins/metabolism , Male , Mice, Transgenic , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Retinal Neoplasms/genetics , Retinal Neoplasms/pathology , Retinoblastoma/genetics , Retinoblastoma/pathology , Retinoblastoma Binding Proteins/genetics , TEA Domain Transcription Factors/metabolism , Ubiquitin-Protein Ligases/genetics , Xenograft Model Antitumor Assays
4.
Dev Dyn ; 243(5): 712-29, 2014 May.
Article in English | MEDLINE | ID: mdl-24339342

ABSTRACT

BACKGROUND: Despite the disease relevance, understanding of human retinal development lags behind that of other species. We compared the kinetics of gene silencing or induction during ganglion cell development in human and murine retina. RESULTS: Induction of POU4F2 (BRN3B) marks ganglion cell commitment, and we detected this factor in S-phase progenitors that had already silenced Cyclin D1 and VSX2 (CHX10). This feature was conserved in human and mouse retina, and the fraction of Pou4f2+ murine progenitors labeled with a 30 min pulse of BrdU matched the fraction of ganglion cells predicted to be born in a half-hour period. Additional analysis of 18 markers revealed many with conserved kinetics, such as the POU4F2 pattern above, as well as the surprising maintenance of "cell cycle" proteins KI67, PCNA, and MCM6 well after terminal mitosis. However, four proteins (TUBB3, MTAP1B, UCHL1, and RBFOX3) showed considerably delayed induction in human relative to mouse retina, and two proteins (ISL1, CALB2) showed opposite kinetics, appearing on either side of terminal mitosis depending on the species. CONCLUSION: With some notable exceptions, human and murine ganglion cell differentiation show similar kinetics, and the data add weight to prior studies supporting the existence of biased ganglion cell progenitors.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle/physiology , Cell Differentiation/physiology , Eye Proteins/metabolism , Retinal Ganglion Cells/metabolism , Stem Cells/metabolism , Animals , Humans , Mice , Retinal Ganglion Cells/cytology , Stem Cells/cytology
5.
Dev Dyn ; 241(10): 1525-44, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22837015

ABSTRACT

BACKGROUND: Knowledge of gene expression kinetics around neuronal cell birth is required to dissect mechanisms underlying progenitor fate. Here, we timed cell cycle and neuronal protein silencing/induction during cell birth in the developing murine retina. RESULTS: The pan-cell cycle markers Pcna and Mcm6 were present in the post-mitotic ganglion cell layer. Although confined to the neuroblastic layer (NBL), 6-7% of Ki67(+) cells lacked six progenitor/cell cycle markers, and expressed neuronal markers. To define protein extinction/induction timing, we defined G2/M length throughout retinogenesis, which was typically 1-2 h, but <10% cells took double this time. BrdU-chase analyses revealed that at E12.5, Tubb3 (Tuj1) appeared at M-phase, followed by Calb2 and Dcx at ~2 h, Elavl2/3/4 at ~4 h, and Map2 at ~6 h after cell birth, and these times extended with embryonic age. Strikingly, Ki67 was not extinguished until up to a day after cell cycle exit, coinciding with exit from the NBL and induction of late markers such as Map1b/Uchl1/Rbfox3. CONCLUSIONS: A minor population of progenitors transits slowly through G2/M and, most importantly, some cell cycle proteins are retained for an unexpectedly long period in post-mitotic neurons. The high-resolution map of cell birth kinetics reported here provides a framework to better define mechanisms that regulate neurogenesis.


Subject(s)
Animals, Newborn/physiology , Cell Cycle Proteins/metabolism , Cell Differentiation/physiology , Gene Expression Regulation, Developmental/physiology , Neurons/metabolism , Retina/cytology , Animals , Bromodeoxyuridine , Doublecortin Protein , G2 Phase Cell Cycle Checkpoints/physiology , Image Processing, Computer-Assisted , Ki-67 Antigen/metabolism , Mice , Microscopy, Confocal , Minichromosome Maintenance Complex Component 6 , Neurons/physiology , Proliferating Cell Nuclear Antigen/metabolism , Retina/growth & development , Time Factors
6.
Nature ; 462(7275): 925-9, 2009 Dec 17.
Article in English | MEDLINE | ID: mdl-20016601

ABSTRACT

The activating E2f transcription factors (E2f1, E2f2 and E2f3) induce transcription and are widely viewed as essential positive cell cycle regulators. Indeed, they drive cells out of quiescence, and the 'cancer cell cycle' in Rb1 null cells is E2f-dependent. Absence of activating E2fs in flies or mammalian fibroblasts causes cell cycle arrest, but this block is alleviated by removing repressive E2f or the tumour suppressor p53, respectively. Thus, whether activating E2fs are indispensable for normal division is an area of debate. Activating E2fs are also well known pro-apoptotic factors, providing a defence against oncogenesis, yet E2f1 can limit irradiation-induced apoptosis. In flies this occurs through repression of hid (also called Wrinkled; Smac/Diablo in mammals). However, in mammals the mechanism is unclear because Smac/Diablo is induced, not repressed, by E2f1, and in keratinocytes survival is promoted indirectly through induction of DNA repair targets. Thus, a direct pro-survival function for E2f1-3 and/or its relevance beyond irradiation has not been established. To address E2f1-3 function in normal cells in vivo we focused on the mouse retina, which is a relatively simple central nervous system component that can be manipulated genetically without compromising viability and has provided considerable insight into development and cancer. Here we show that unlike fibroblasts, E2f1-3 null retinal progenitor cells or activated Müller glia can divide. We attribute this effect to functional interchangeability with Mycn. However, loss of activating E2fs caused downregulation of the p53 deacetylase Sirt1, p53 hyperacetylation and elevated apoptosis, establishing a novel E2f-Sirt1-p53 survival axis in vivo. Thus, activating E2fs are not universally required for normal mammalian cell division, but have an unexpected pro-survival role in development.


Subject(s)
Apoptosis , E2F Transcription Factors/deficiency , Retina/cytology , Retina/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Acetylation , Animals , Cell Division , Cell Survival , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/metabolism , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , E2F1 Transcription Factor/deficiency , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , E2F2 Transcription Factor/deficiency , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism , E2F3 Transcription Factor/deficiency , E2F3 Transcription Factor/genetics , E2F3 Transcription Factor/metabolism , Fibroblasts , Mice , Mice, Knockout , Neuroglia/cytology , Neuroglia/metabolism , Sirtuin 1/metabolism , Tumor Suppressor Protein p53/metabolism
7.
PLoS One ; 4(10): e7507, 2009 Oct 19.
Article in English | MEDLINE | ID: mdl-19838301

ABSTRACT

BACKGROUND: Optical coherence tomography (OCT) is a novel method of retinal in vivo imaging. In this study, we assessed the potential of OCT to yield histology-analogue sections in mouse models of retinal degeneration. METHODOLOGY/PRINCIPAL FINDINGS: We achieved to adapt a commercial 3(rd) generation OCT system to obtain and quantify high-resolution morphological sections of the mouse retina which so far required in vitro histology. OCT and histology were compared in models with developmental defects, light damage, and inherited retinal degenerations. In conditional knockout mice deficient in retinal retinoblastoma protein Rb, the gradient of Cre expression from center to periphery, leading to a gradual reduction of retinal thickness, was clearly visible and well topographically quantifiable. In Nrl knockout mice, the layer involvement in the formation of rosette-like structures was similarly clear as in histology. OCT examination of focal light damage, well demarcated by the autofluorescence pattern, revealed a practically complete loss of photoreceptors with preservation of inner retinal layers, but also more subtle changes like edema formation. In Crb1 knockout mice (a model for Leber's congenital amaurosis), retinal vessels slipping through the outer nuclear layer towards the retinal pigment epithelium (RPE) due to the lack of adhesion in the subapical region of the photoreceptor inner segments could be well identified. CONCLUSIONS/SIGNIFICANCE: We found that with the OCT we were able to detect and analyze a wide range of mouse retinal pathology, and the results compared well to histological sections. In addition, the technique allows to follow individual animals over time, thereby reducing the numbers of study animals needed, and to assess dynamic processes like edema formation. The results clearly indicate that OCT has the potential to revolutionize the future design of respective short- and long-term studies, as well as the preclinical assessment of therapeutic strategies.


Subject(s)
Retina/metabolism , Retinal Degeneration/metabolism , Tomography, Optical Coherence/methods , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Eye Proteins/genetics , Female , Lasers , Light , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Ophthalmoscopy/methods , Retinoblastoma Protein/genetics
8.
Viral Immunol ; 20(3): 495-502, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17931120

ABSTRACT

To warrant potential clinical testing, the equine anti-severe acute respiratory syndrome coronavirus (SARS-CoV) F(ab')(2) requires evaluation in as many animal models as possible. In this study, we established a new animal model, the Chinese hamster, susceptible to SARS-CoV infection. SARS-CoV could propagate effectively and sustain high levels for 1 wk in animal lungs. All animals were protected from SARS-CoV infection in preventive settings. Further, when used therapeutically this antibody led to an approximately 4-log(10) decrease in viral burden in infected animal lungs. The pathological changes in lungs correlated closely with the dose of antibody administered. The excellent preventive and therapeutic roles of equine anti-SARS-CoV F(ab')(2) in several animal models, including the novel Chinese hamster model described in this study, have provided exciting data concerning its potential clinical study.


Subject(s)
Antibodies, Viral/therapeutic use , Immunoglobulin Fab Fragments/therapeutic use , Severe Acute Respiratory Syndrome/prevention & control , Animals , Cricetinae , Cricetulus , Disease Models, Animal , Female , Horses/immunology , Lung/pathology , Lung/virology , Neutralization Tests , Severe acute respiratory syndrome-related coronavirus/isolation & purification , Severe Acute Respiratory Syndrome/immunology
9.
PLoS Biol ; 5(7): e179, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17608565

ABSTRACT

It has long been known that loss of the retinoblastoma protein (Rb) perturbs neural differentiation, but the underlying mechanism has never been solved. Rb absence impairs cell cycle exit and triggers death of some neurons, so differentiation defects may well be indirect. Indeed, we show that abnormalities in both differentiation and light-evoked electrophysiological responses in Rb-deficient retinal cells are rescued when ectopic division and apoptosis are blocked specifically by deleting E2f transcription factor (E2f) 1. However, comprehensive cell-type analysis of the rescued double-null retina exposed cell-cycle-independent differentiation defects specifically in starburst amacrine cells (SACs), cholinergic interneurons critical in direction selectivity and developmentally important rhythmic bursts. Typically, Rb is thought to block division by repressing E2fs, but to promote differentiation by potentiating tissue-specific factors. Remarkably, however, Rb promotes SAC differentiation by inhibiting E2f3 activity. Two E2f3 isoforms exist, and we find both in the developing retina, although intriguingly they show distinct subcellular distribution. E2f3b is thought to mediate Rb function in quiescent cells. However, in what is to our knowledge the first work to dissect E2f isoform function in vivo we show that Rb promotes SAC differentiation through E2f3a. These data reveal a mechanism through which Rb regulates neural differentiation directly, and, unexpectedly, it involves inhibition of E2f3a, not potentiation of tissue-specific factors.


Subject(s)
Amacrine Cells/cytology , Amacrine Cells/metabolism , E2F3 Transcription Factor/metabolism , Retinoblastoma Protein/metabolism , Animals , Apoptosis , Base Sequence , Cell Cycle , Cell Differentiation/physiology , DNA Primers/genetics , E2F1 Transcription Factor/deficiency , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , E2F2 Transcription Factor/deficiency , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/metabolism , E2F3 Transcription Factor/deficiency , E2F3 Transcription Factor/genetics , Female , Genes, Retinoblastoma , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Biological , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/genetics
10.
Viral Immunol ; 20(1): 197-205, 2007.
Article in English | MEDLINE | ID: mdl-17425434

ABSTRACT

Equine anti-severe acute respiratory syndrome-associated coronavirus F(ab')(2) has been verified to protect mice from infection with severe acute respiratory syndrome-associated coronavirus (SARS-CoV). However, before potential clinical application, the antibody needs to be tested in as many animal models as possible to ensure its safety and efficiency. In this study, after verification by various methods that the golden Syrian hamster constitutes a model susceptible to SARS-CoV infection, we confirmed that the antibody could protect animals completely from SARS-CoV infection in the preventive setting. More importantly, the antibody could reduce viral titers or copies by approximately 10(3)- to 10(4)-fold in animal lung after virus exposure, compared with negative control. These data provide further evidence to warrant clinical studies of this antibody in the treatment and prevention of SARS.


Subject(s)
Antibodies, Viral/immunology , Immunoglobulin Fab Fragments/immunology , Severe Acute Respiratory Syndrome/prevention & control , Administration, Intranasal , Animals , Cricetinae , Horses , Mesocricetus , Neutralization Tests , Polymerase Chain Reaction
11.
Curr Mol Med ; 6(7): 759-81, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17100602

ABSTRACT

The RB gene was discovered 20 years ago because of its role in the childhood eye cancer retinoblastoma. However, surprisingly little progress was made in defining the role of RB protein in the retina. In the last two years, new models exploiting conditional deletion of the mouse Rb gene have altered this picture radically. These models provide insight into the first Rb function, the cell of origin of retinoblastoma, the window during which Rb acts, distinct cell-specific defenses against Rb loss, the number and type of post-Rb lesions required for transformation, why pediatric tumors exist, the controversial role of the p53 pathway in retinoblastoma, and the reason why the disease is virtually unique to humans. Two years have dramatically improved our understanding of Rb function in the tissue that gave us this important tumor suppressor.


Subject(s)
Cell Transformation, Neoplastic , Disease Models, Animal , Retinoblastoma Protein/metabolism , Animals , Disease Progression , Humans , Retinoblastoma Protein/genetics
12.
Mol Cell Neurosci ; 32(3): 274-82, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16815712

ABSTRACT

The morphogen Sonic hedgehog (Shh) is expressed by the projection neurons of the retina, retinal ganglion cells (RGCs) and promotes retinal precursor cell (RPC) proliferation. To distinguish between direct and indirect effects of Hedgehog (Hh) pathway activation in the perinatal mouse retina, we followed the fate of cells that expressed a constitutively active allele of Smoothened (SMO-M2), the signal transduction component of the Hh pathway. SMO-M2 expression promoted a cell-autonomous increase in CyclinD1 expression and RPC proliferation and promoted the development of cells with an inner nuclear layer identity. SMO-M2 expression also inhibited rhodopsin expression in uninfected cells, thus highlighting an unexpected non-cell autonomous effect of Hh pathway activation on photoreceptor development.


Subject(s)
Retina/metabolism , Signal Transduction/physiology , Trans-Activators/metabolism , Animals , Cells, Cultured , Hedgehog Proteins , Humans , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , NIH 3T3 Cells , Receptors, G-Protein-Coupled/biosynthesis , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Retina/growth & development , Retina/physiology , Smoothened Receptor , Trans-Activators/genetics , Trans-Activators/physiology
13.
Proc Natl Acad Sci U S A ; 103(13): 4988-93, 2006 Mar 28.
Article in English | MEDLINE | ID: mdl-16547132

ABSTRACT

In the Chx10-null ocular retardation (or(J)) mouse, retinal progenitor cell (RPC) proliferation is impaired, and bipolar neurons, a late born cell type, fail to differentiate. It is unclear whether Chx10 is required to maintain proliferation throughout retinogenesis or whether the bipolar cell defect is an indirect effect of growth arrest. We show that Chx10 is dispensable for late-stage RPC proliferation but is essential to promote bipolar cell genesis in place of rods. Ectopic Chx10 expression drove bipolar instead of rod cell differentiation without affecting division. Converting Chx10 to an activator impaired bipolar cell differentiation, implying that repression is important for Chx10 activity. In the Chx10 null or(J) retina, only a small fraction of cells expressing mutated Chx10 mRNA were rods, but this fraction increased after p27(Kip1) inactivation, which partially rescues proliferation. Most significantly, acute Chx10 knockdown in the postnatal retina promoted rods in place of bipolar neurons without affecting division. Thus, Chx10 directly controls bipolar cell genesis by inhibiting rod differentiation independent of its temporally limited early effect on RPC proliferation.


Subject(s)
Cell Differentiation , Homeodomain Proteins/metabolism , Photoreceptor Cells/cytology , Photoreceptor Cells/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Animals, Newborn , Cell Polarity , Cell Proliferation , Homeodomain Proteins/genetics , Mice , Mice, Knockout , RNA, Messenger/genetics , Rats , Transcription Factors/deficiency , Transcription Factors/genetics
14.
Biochem Biophys Res Commun ; 316(4): 978-83, 2004 Apr 16.
Article in English | MEDLINE | ID: mdl-15044080

ABSTRACT

A well-established feature of the translation initiation region, which attracts the ribosomes to the prokaryotic mRNAs, is a purine rich area called Shine/Dalgarno sequence (SD). There are examples of various other sequences, which despite having no similarity to an SD sequence are capable of enhancing and/or initiating translation. The mechanisms by which these sequences affect translation remain unclear, but a base pairing between mRNA and 16S ribosomal RNA (rRNA) is proposed to be the likely mechanism. In this study, using a computational approach, we identified a non-SD signal found specifically in the translation initiation regions of Escherichia coli mRNAs, which contain super strong SD sequences. Nine of the 11 E. coli translation initiation regions, which were previously identified for having super strong SD sequences, also contained six or more nucleotides complementary to box-17 on the 16S rRNA (nucleotides 418-554). Mutational analyses of those initiation sequences indicated that when complementarity to box-17 was eliminated, the efficiency of the examined sequences to mediate the translation of chloramphenicol acetyltransferase (CAT) mRNA was reduced. The results suggest that mRNA sequences with complementarity to box-17 of 16S rRNA may function as enhancers for translation in E. coli.


Subject(s)
Base Pairing/genetics , Escherichia coli/genetics , Gene Expression Profiling/methods , Protein Biosynthesis/genetics , RNA, Complementary/genetics , RNA, Messenger/genetics , RNA, Ribosomal, 16S/genetics , Sequence Analysis, RNA/methods , Gene Expression Regulation, Bacterial/genetics , Sequence Alignment/methods , Sequence Homology, Nucleic Acid
15.
Dev Neurosci ; 26(5-6): 417-34, 2004.
Article in English | MEDLINE | ID: mdl-15855771

ABSTRACT

The Rb gene was isolated almost 20 years ago, but fundamental questions regarding its role in retinal development and retinoblastoma remain. What is the normal function of RB protein in retinogenesis? What is the cell-of-origin of retinoblastoma? Why do retinoblastoma tumors have recurrent genetic lesions other than Rb inactivation? Why is retinoblastoma not induced by defects in cell cycle regulators other than Rb? Why is the retina so sensitive to Rb loss? Recently developed conditional Rb knockout models provide new insight into some of these issues. The data suggest that RB protein may not control the rate of progenitor division, but is critical for cell cycle exit when dividing retinal progenitors differentiate into postmitotic transition cells. This finding focuses attention on the ectopically dividing transition cell, rather than the progenitor, as the cell-of-origin. Cell-specific analyses in the RB-deficient retina reveal that ectopically dividing photoreceptors, bipolar and ganglion cells die, but amacrine, horizontal and Muller cells survive and stop dividing when they terminally differentiate. Rare amacrine transition cells escape cell cycle exit and generate tumors. These data suggest that post-Rb mutations are required to overcome growth arrest associated with terminal differentiation, rather than apoptosis as previously suggested. To explain why perturbing cell cycle regulators other than RB does not initiate retinoblastoma, we speculate that mutations in other components of the RB pathway perturb cell cycle arrest, but only RB loss triggers genome instability in retinal transition cells, which may be critical to facilitate post-Rb mutations necessary for transformation. Cell-specific differences in the effect of Rb loss on genome stability may contribute to the tremendous sensitivity of retinal transition cells to tumorigenesis. The new mouse models of retinoblastoma will be invaluable for testing these possibilities.


Subject(s)
Cell Cycle Proteins/metabolism , Retina/embryology , Retinal Neoplasms/metabolism , Retinoblastoma Protein/metabolism , Retinoblastoma/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Differentiation/physiology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Genomic Instability/physiology , Humans , Mice , Mice, Knockout , Mutation/physiology , Retina/cytology , Retina/metabolism , Retinal Neoplasms/genetics , Retinal Neoplasms/physiopathology , Retinoblastoma/genetics , Retinoblastoma/physiopathology , Retinoblastoma Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL