Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters











Publication year range
1.
Curr Opin Behav Sci ; 562024 Apr.
Article in English | MEDLINE | ID: mdl-39130377

ABSTRACT

Repetitive negative thinking (RNT) is a transdiagnostic construct that encompasses rumination and worry, yet what precisely is shared between rumination and worry is unclear. To clarify this, we develop a meta-control account of RNT. Meta-control refers to the reinforcement and control of mental behavior via similar computations as reinforce and control motor behavior. We propose rumination and worry are coarse terms for failure in meta-control, just as tripping and falling are coarse terms for failure in motor control. We delineate four meta-control stages and risk factors increasing the chance of failure at each, including open-ended thoughts (stage 1), individual differences influencing subgoal execution (stage 2) and switching (stage 3), and challenges inherent to learning adaptive mental behavior (stage 4). Distinguishing these stages therefore elucidates diverse processes that lead to the same behavior of excessive RNT. Our account also subsumes prominent clinical accounts of RNT into a computational cognitive neuroscience framework.

2.
J Psychopathol Clin Sci ; 133(5): 413-426, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38815082

ABSTRACT

Many psychotherapies aim to help people replace maladaptive mental behaviors (such as those leading to unproductive worry) with more adaptive ones (such as those leading to active problem solving). Yet, little is known empirically about how challenging it is to learn adaptive mental behaviors. Mental behaviors entail taking mental operations and thus may be more challenging to perform than motor actions; this challenge may enhance or impair learning. In particular, challenge when learning is often desirable because it improves retention. Yet, it is also plausible that the necessity of carrying out mental operations interferes with learning the expected values of mental actions by impeding credit assignment: the process of updating an action's value after reinforcement. Then, it may be more challenging not only to perform-but also to learn the consequences of-mental (vs. motor) behaviors. We designed a task to assess learning to take adaptive mental versus motor actions via matched probabilistic feedback. In two experiments (N = 300), most participants found it more difficult to learn to select optimal mental (vs. motor) actions, as evident in worse accuracy not only in a learning but also test (retention) phase. Computational modeling traced this impairment to an indicator of worse credit assignment (impaired construction and maintenance of expected values) when learning mental actions, accounting for worse accuracy in the learning and retention phases. The results suggest that people have particular difficulty learning adaptive mental behavior and pave the way for novel interventions to scaffold credit assignment and promote adaptive thinking. (PsycInfo Database Record (c) 2024 APA, all rights reserved).


Subject(s)
Adaptation, Psychological , Learning , Humans , Learning/physiology , Adult , Male , Female , Young Adult , Reinforcement, Psychology
4.
Nat Commun ; 14(1): 8477, 2023 Dec 20.
Article in English | MEDLINE | ID: mdl-38123561

ABSTRACT

Following acute retinal damage, zebrafish possess the ability to regenerate all neuronal subtypes through Müller glia (MG) reprogramming and asymmetric cell division that produces a multipotent Müller glia-derived neuronal progenitor cell (MGPC). This raises three key questions. First, do MG reprogram to a developmental retinal progenitor cell (RPC) state? Second, to what extent does regeneration recapitulate retinal development? And finally, does loss of different retinal cell subtypes induce unique MG regeneration responses? We examined these questions by performing single-nuclear and single-cell RNA-Seq and ATAC-Seq in both developing and regenerating retinas. Here we show that injury induces MG to reprogram to a state similar to late-stage RPCs. However, there are major transcriptional differences between MGPCs and RPCs, as well as major transcriptional differences between activated MG and MGPCs when different retinal cell subtypes are damaged. Validation of candidate genes confirmed that loss of different subtypes induces differences in transcription factor gene expression and regeneration outcomes.


Subject(s)
Gene Regulatory Networks , Zebrafish , Animals , Zebrafish/genetics , Retina/metabolism , Neurogenesis/genetics , Neuroglia/metabolism , Cell Proliferation/physiology , Ependymoglial Cells/metabolism
5.
bioRxiv ; 2023 Aug 08.
Article in English | MEDLINE | ID: mdl-37609307

ABSTRACT

Following acute retinal damage, zebrafish possess the ability to regenerate all neuronal subtypes. This regeneration requires Müller glia (MG) to reprogram and divide asymmetrically to produce a multipotent Müller glia-derived neuronal progenitor cell (MGPC). This raises three key questions. First, does loss of different retinal cell subtypes induce unique MG regeneration responses? Second, do MG reprogram to a developmental retinal progenitor cell state? And finally, to what extent does regeneration recapitulate retinal development? We examined these questions by performing single-nuclear and single-cell RNA-Seq and ATAC-Seq in both developing and regenerating retinas. While MG reprogram to a state similar to late-stage retinal progenitors in developing retinas, there are transcriptional differences between reprogrammed MG/MGPCs and late progenitors, as well as reprogrammed MG in outer and inner retinal damage models. Validation of candidate genes confirmed that loss of different subtypes induces differences in transcription factor gene expression and regeneration outcomes. This work identifies major differences between gene regulatory networks activated following the selective loss of different subtypes of retina neurons, as well as between retinal regeneration and development.

6.
Cogn Affect Behav Neurosci ; 23(1): 171-189, 2023 02.
Article in English | MEDLINE | ID: mdl-36168080

ABSTRACT

Cognitive theories of depression, and mindfulness theories of well-being, converge on the notion that self-judgment plays a critical role in mental health. However, these theories have rarely been tested via tasks and computational modeling analyses that can disentangle the information processes operative in self-judgments. We applied a drift-diffusion computational model to the self-referential encoding task (SRET) collected before and after an 8-week mindfulness intervention (n = 96). A drift-rate regression parameter representing positive-relative to negative-self-referential judgment strength positively related to mindful awareness and inversely related to depression, both at baseline and over time; however, this parameter did not significantly relate to the interaction between mindful awareness and nonjudgmentalness. At the level of individual depression symptoms, at baseline, a spectrum of symptoms (inversely) correlated with the drift-rate regression parameter, suggesting that many distinct depression symptoms relate to valenced self-judgment between subjects. By contrast, over the intervention, changes in only a smaller subset of anhedonia-related depression symptoms showed substantial relationships with this parameter. Both behavioral and model-derived measures showed modest split-half and test-retest correlations. Results support cognitive theories that implicate self-judgment in depression and mindfulness theories, which imply that mindful awareness should lead to more positive self-views.


Subject(s)
Depression , Mindfulness , Humans , Judgment , Cognition , Computer Simulation
7.
Mol Neurobiol ; 59(5): 2910-2931, 2022 May.
Article in English | MEDLINE | ID: mdl-35246819

ABSTRACT

In mammals, photoreceptor loss causes permanent blindness, but in zebrafish (Danio rerio), photoreceptor loss reprograms Müller glia to function as stem cells, producing progenitors that regenerate photoreceptors. MicroRNAs (miRNAs) regulate CNS neurogenesis, but the roles of miRNAs in injury-induced neuronal regeneration are largely unknown. In the embryonic zebrafish retina, miR-18a regulates photoreceptor differentiation. The purpose of the current study was to determine, in zebrafish, the function of miR-18a during injury-induced photoreceptor regeneration. RT-qPCR, in situ hybridization, and immunohistochemistry showed that miR-18a expression increases throughout the retina between 1 and 5 days post-injury (dpi). To test miR-18a function during photoreceptor regeneration, we used homozygous miR-18a mutants (miR-18ami5012), and knocked down miR-18a with morpholino oligonucleotides. During photoreceptor regeneration, miR-18ami5012 retinas have fewer mature photoreceptors than WT at 7 and 10 dpi, but there is no difference at 14 dpi, indicating that photoreceptor regeneration is delayed. Labeling dividing cells with 5-bromo-2'-deoxyuridine (BrdU) showed that at 7 and 10 dpi, there are excess dividing progenitors in both mutants and morphants, indicating that miR-18a negatively regulates injury-induced proliferation. Tracing 5-ethynyl-2'-deoxyuridine (EdU) and BrdU-labeled cells showed that in miR-18ami5012 retinas excess progenitors migrate to other retinal layers in addition to the photoreceptor layer. Inflammation is critical for photoreceptor regeneration, and RT-qPCR showed that in miR-18ami5012 retinas, inflammatory gene expression and microglia activation are prolonged. Suppressing inflammation with dexamethasone rescues the miR-18ami5012 phenotype. Together, these data show that in the injured zebrafish retina, disruption of miR-18a alters proliferation, inflammation, the microglia/macrophage response, and the timing of photoreceptor regeneration.


Subject(s)
MicroRNAs , Zebrafish , Animals , Bromodeoxyuridine/metabolism , Cell Proliferation/physiology , Inflammation/metabolism , Kinetics , Macrophages , Mammals , MicroRNAs/genetics , MicroRNAs/metabolism , Microglia , Retina/metabolism , Zebrafish/metabolism
8.
Development ; 149(4)2022 02 15.
Article in English | MEDLINE | ID: mdl-35088848

ABSTRACT

Endothelial cells emerge from the atrioventricular canal to form coronary blood vessels in juvenile zebrafish hearts. We find that pdgfrb is first expressed in the epicardium around the atrioventricular canal and later becomes localized mainly in the mural cells. pdgfrb mutant fish show severe defects in mural cell recruitment and coronary vessel development. Single-cell RNA sequencing analyses identified pdgfrb+ cells as epicardium-derived cells (EPDCs) and mural cells. Mural cells associated with coronary arteries also express cxcl12b and smooth muscle cell markers. Interestingly, these mural cells remain associated with coronary arteries even in the absence of Pdgfrß, although smooth muscle gene expression is downregulated. We find that pdgfrb expression dynamically changes in EPDCs of regenerating hearts. Differential gene expression analyses of pdgfrb+ EPDCs and mural cells suggest that they express genes that are important for regeneration after heart injuries. mdka was identified as a highly upregulated gene in pdgfrb+ cells during heart regeneration. However, pdgfrb but not mdka mutants show defects in heart regeneration after amputation. Our results demonstrate that heterogeneous pdgfrb+ cells are essential for coronary development and heart regeneration.


Subject(s)
Coronary Vessels/growth & development , Coronary Vessels/metabolism , Heart/physiology , Organogenesis/physiology , Receptor, Platelet-Derived Growth Factor beta/metabolism , Regeneration/physiology , Animals , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental/physiology , Myocytes, Smooth Muscle/metabolism , Pericardium/metabolism , Zebrafish/metabolism , Zebrafish/physiology
9.
Annu Rev Psychol ; 73: 243-270, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34579545

ABSTRACT

Why has computational psychiatry yet to influence routine clinical practice? One reason may be that it has neglected context and temporal dynamics in the models of certain mental health problems. We develop three heuristics for estimating whether time and context are important to a mental health problem: Is it characterized by a core neurobiological mechanism? Does it follow a straightforward natural trajectory? And is intentional mental content peripheral to the problem? For many problems the answers are no, suggesting that modeling time and context is critical. We review computational psychiatry advances toward this end, including modeling state variation, using domain-specific stimuli, and interpreting differences in context. We discuss complementary network and complex systems approaches. Novel methods and unification with adjacent fields may inspire a new generation of computational psychiatry.


Subject(s)
Mental Disorders , Psychiatry , Humans , Mental Health
10.
Schizophr Res Cogn ; 25: 100196, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33996517

ABSTRACT

People with schizophrenia (SZ) process emotions less accurately than do healthy comparators (HC), and emotion recognition have expanded beyond accuracy to performance variables like reaction time (RT) and confidence. These domains are typically evaluated independently, but complex inter-relationships can be evaluated through machine learning at an item-by-item level. Using a mix of ranking and machine learning tools, we investigated item-by-item discrimination of facial affect with two emotion recognition tests (BLERT and ER-40) between SZ and HC. The best performing multi-domain model for ER40 had a large effect size in differentiating SZ and HC (d = 1.24) compared to a standard comparison of accuracy alone (d = 0.48); smaller increments in effect sizes were evident for the BLERT (d = 0.87 vs. d = 0.58). Almost half of the selected items were confidence ratings. Within SZ, machine learning models with ER40 (generally accuracy and reaction time) items predicted severity of depression and overconfidence in social cognitive ability, but not psychotic symptoms. Pending independent replication, the results support machine learning, and the inclusion of confidence ratings, in characterizing the social cognitive deficits in SZ. This moderate-sized study (n = 372) included subjects with schizophrenia (SZ, n = 218) and healthy controls (HC, n = 154).

11.
Cells ; 10(4)2021 04 01.
Article in English | MEDLINE | ID: mdl-33916186

ABSTRACT

The ability to regenerate tissues varies between species and between tissues within a species. Mammals have a limited ability to regenerate tissues, whereas zebrafish possess the ability to regenerate almost all tissues and organs, including fin, heart, kidney, brain, and retina. In the zebrafish brain, injury and cell death activate complex signaling networks that stimulate radial glia to reprogram into neural stem-like cells that repair the injury. In the retina, a popular model for investigating neuronal regeneration, Müller glia, radial glia unique to the retina, reprogram into stem-like cells and undergo a single asymmetric division to generate multi-potent retinal progenitors. Müller glia-derived progenitors then divide rapidly, numerically matching the magnitude of the cell death, and differentiate into the ablated neurons. Emerging evidence reveals that inflammation plays an essential role in this multi-step process of retinal regeneration. This review summarizes the current knowledge of the inflammatory events during retinal regeneration and highlights the mechanisms whereby inflammatory molecules regulate the quiescence and division of Müller glia, the proliferation of Müller glia-derived progenitors and the survival of regenerated neurons.


Subject(s)
Inflammation/pathology , Regeneration/physiology , Retina/physiopathology , Zebrafish/physiology , Animals , Cellular Reprogramming , Ependymoglial Cells/pathology , Neurogenesis
12.
PLoS One ; 15(6): e0232308, 2020.
Article in English | MEDLINE | ID: mdl-32530962

ABSTRACT

Zebrafish have the ability to regenerate damaged cells and tissues by activating quiescent stem and progenitor cells or reprogramming differentiated cells into regeneration-competent precursors. Proliferation among the cells that will functionally restore injured tissues is a fundamental biological process underlying regeneration. Midkine-a is a cytokine growth factor, whose expression is strongly induced by injury in a variety of tissues across a range of vertebrate classes. Using a zebrafish Midkine-a loss of function mutant, we evaluated regeneration of caudal fin, extraocular muscle and retinal neurons to investigate the function of Midkine-a during epimorphic regeneration. In wildtype zebrafish, injury among these tissues induces robust proliferation and rapid regeneration. In Midkine-a mutants, the initial proliferation in each of these tissues is significantly diminished or absent. Regeneration of the caudal fin and extraocular muscle is delayed; regeneration of the retina is nearly completely absent. These data demonstrate that Midkine-a is universally required in the signaling pathways that convert tissue injury into the initial burst of cell proliferation. Further, these data highlight differences in the molecular mechanisms that regulate epimorphic regeneration in zebrafish.


Subject(s)
Midkine/metabolism , Regeneration/physiology , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animal Fins/physiology , Animals , Animals, Genetically Modified/metabolism , Cell Differentiation , Cell Proliferation , Midkine/genetics , Mutagenesis , Neuroglia/cytology , Neuroglia/metabolism , Oculomotor Muscles/physiology , Retinal Neurons/physiology , Zebrafish Proteins/genetics
13.
Annu Rev Vis Sci ; 6: 171-193, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32343929

ABSTRACT

In humans, various genetic defects or age-related diseases, such as diabetic retinopathies, glaucoma, and macular degeneration, cause the death of retinal neurons and profound vision loss. One approach to treating these diseases is to utilize stem and progenitor cells to replace neurons in situ, with the expectation that new neurons will create new synaptic circuits or integrate into existing ones. Reprogramming non-neuronal cells in vivo into stem or progenitor cells is one strategy for replacing lost neurons. Zebrafish have become a valuable model for investigating cellular reprogramming and retinal regeneration. This review summarizes our current knowledge regarding spontaneous reprogramming of Müller glia in zebrafish and compares this knowledge to research efforts directed toward reprogramming Müller glia in mammals. Intensive research using these animal models has revealed shared molecular mechanisms that make Müller glia attractive targets for cellular reprogramming and highlighted the potential for curing degenerative retinal diseases from intrinsic cellular sources.


Subject(s)
Ependymoglial Cells/physiology , Nerve Regeneration/physiology , Retinal Neurons/physiology , Animals , Animals, Genetically Modified , Cell Differentiation , DNA Methylation , Epigenomics , Humans , Receptors, Notch/metabolism , Retinal Neurons/cytology , Signal Transduction , Stem Cells , Zebrafish
14.
Glia ; 68(7): 1445-1465, 2020 07.
Article in English | MEDLINE | ID: mdl-32034934

ABSTRACT

Brain injury activates complex inflammatory signals in dying neurons, surviving neurons, and glia. Here, we establish that inflammation regulates the regeneration of photoreceptors in the zebrafish retina and determine the cellular expression and function of the inflammatory protease, matrix metalloproteinase 9 (Mmp-9), during this regenerative neurogenesis. Following photoreceptor ablation, anti-inflammatory treatment suppresses the number of injury-induced progenitors and regenerated photoreceptors. Upon photoreceptor injury, mmp-9 is induced in Müller glia and Müller glia-derived photoreceptor progenitors. Deleting mmp-9 results in over production of injury-induced progenitors and regenerated photoreceptors, but over time the absence of Mmp-9 compromises the survival of the regenerated cones. At all time-points studied, the levels of tnf-α are significantly elevated in mutant retinas. Anti-inflammatory treatment in mutants rescues the defects in cone survival. These data provide a link between injury-induced inflammation in the vertebrate CNS, Mmp-9 function during neuronal regeneration and the requirement of Mmp-9 for the survival of regenerated cones.


Subject(s)
Inflammation/metabolism , Matrix Metalloproteinase 9/metabolism , Nerve Regeneration/physiology , Regeneration/physiology , Animals , Animals, Genetically Modified , Cell Differentiation/physiology , Cell Proliferation/physiology , Neuroglia/metabolism , Retina/metabolism , Retinal Rod Photoreceptor Cells/physiology , Stem Cells/physiology , Zebrafish
15.
J Neurosci ; 40(6): 1232-1247, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31882403

ABSTRACT

In the retina of zebrafish, Müller glia have the ability to reprogram into stem cells capable of regenerating all classes of retinal neurons and restoring visual function. Understanding the cellular and molecular mechanisms controlling the stem cell properties of Müller glia in zebrafish may provide cues to unlock the regenerative potential in the mammalian nervous system. Midkine is a cytokine/growth factor with multiple roles in neural development, tissue repair, and disease. In midkine-a loss-of-function mutants of both sexes, Müller glia initiate the appropriate reprogramming response to photoreceptor death by increasing expression of stem cell-associated genes, and entering the G1 phase of the cell cycle. However, transition from G1 to S phase is blocked in the absence of Midkine-a, resulting in significantly reduced proliferation and selective failure to regenerate cone photoreceptors. Failing to progress through the cell cycle, Müller glia undergo reactive gliosis, a pathological hallmark in the injured CNS of mammals. Finally, we determined that the Midkine-a receptor, anaplastic lymphoma kinase, is upstream of the HLH regulatory protein, Id2a, and of the retinoblastoma gene, p130, which regulates progression through the cell cycle. These results demonstrate that Midkine-a functions as a core component of the mechanisms that regulate proliferation of stem cells in the injured CNS.SIGNIFICANCE STATEMENT The death of retinal neurons and photoreceptors is a leading cause of vision loss. Regenerating retinal neurons is a therapeutic goal. Zebrafish can regenerate retinal neurons from intrinsic stem cells, Müller glia, and are a powerful model to understand how stem cells might be used therapeutically. Midkine-a, an injury-induced growth factor/cytokine that is expressed by Müller glia following neuronal death, is required for Müller glia to progress through the cell cycle. The absence of Midkine-a suspends proliferation and neuronal regeneration. With cell cycle progression stalled, Müller glia undergo reactive gliosis, a pathological hallmark of the mammalian retina. This work provides a unique insight into mechanisms that control the cell cycle during neuronal regeneration.


Subject(s)
Cell Dedifferentiation/physiology , Cellular Reprogramming/physiology , Midkine/metabolism , Nerve Regeneration/physiology , Neuroglia , Retina , Animals , Animals, Genetically Modified , Cell Cycle/physiology , Cell Proliferation/physiology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Retina/cytology , Retina/metabolism , Zebrafish , Zebrafish Proteins/metabolism
16.
Bio Protoc ; 10(24): e3848, 2020 Dec 20.
Article in English | MEDLINE | ID: mdl-33659497

ABSTRACT

Immunohistochemistry is a widely used technique to examine the expression and subcellular localization of proteins. This technique relies on the specificity of antibodies and requires adequate penetration of antibodies into tissues. The latter is especially challenging for thick specimens, such as embryos and other whole-mount preparations. Here we describe an improved method of immunohistochemistry for retinal whole-mount preparations. We report that a cocktail of three reagents, Triton X-100, Tween-20, and DMSO, in blocking and antibody dilution buffers strongly enhances immunolabeling in whole-mount retinas from adult zebrafish. In addition, we establish that in whole retinal tissues, a classic epitope retrieval method, based on citrate buffer, is effective for immunolabeling membrane-associated proteins. Overall, this simple modification allows precise and reproducible immunolabeling of proteins in retinal whole-mounts.

17.
Dev Neurobiol ; 79(2): 202-219, 2019 02.
Article in English | MEDLINE | ID: mdl-30615274

ABSTRACT

During embryonic retinal development, six types of retinal neurons are generated from multipotent progenitors in a strict spatiotemporal pattern. This pattern requires cell cycle exit (i.e. neurogenesis) and differentiation to be precisely regulated in a lineage-specific manner. In zebrafish, the bHLH transcription factor NeuroD governs photoreceptor genesis through Notch signaling but also governs photoreceptor differentiation though distinct mechanisms that are currently unknown. Also unknown are the mechanisms that regulate NeuroD and the spatiotemporal pattern of photoreceptor development. Members of the miR-17-92 microRNA cluster regulate CNS neurogenesis, and a member of this cluster, miR-18a, is predicted to target neuroD mRNA. The purpose of this study was to determine if, in the developing zebrafish retina, miR-18a regulates NeuroD and if it plays a role in photoreceptor development. Quantitative RT-PCR showed that, of the three miR-18 family members (miR-18a, b, and c), miR-18a expression most closely parallels neuroD expression. Morpholino oligonucleotides and CRISPR/Cas9 gene editing were used for miR-18a loss-of-function (LOF) and both resulted in larvae with more mature photoreceptors at 70 hpf without affecting cell proliferation. Western blot showed that miR-18a LOF increases NeuroD protein levels and in vitro dual luciferase assay showed that miR-18a directly interacts with the 3' UTR of neuroD. Finally, tgif1 mutants have increased miR-18a expression, less NeuroD protein and fewer mature photoreceptors, and the photoreceptor deficiency is rescued by miR-18a knockdown. Together, these results show that, independent of neurogenesis, miR-18a regulates the timing of photoreceptor differentiation and indicate that this occurs through post-transcriptional regulation of NeuroD.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , MicroRNAs/genetics , Nerve Tissue Proteins/metabolism , Retina/growth & development , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation, Developmental/genetics , MicroRNAs/metabolism , Neurogenesis/genetics , Transcription Factors/metabolism , Zebrafish/metabolism
18.
Dev Neurobiol ; 77(9): 1114-1129, 2017 09.
Article in English | MEDLINE | ID: mdl-28380680

ABSTRACT

We evaluated the expression and function of the microglia-specific growth factor, Progranulin-a (Pgrn-a) during developmental neurogenesis in the embryonic retina of zebrafish. At 24 hpf pgrn-a is expressed throughout the forebrain, but by 48 hpf pgrn-a is exclusively expressed by microglia and/or microglial precursors within the brain and retina. Knockdown of Pgrn-a does not alter the onset of neurogenic programs or increase cell death, however, in its absence, neurogenesis is significantly delayed-retinal progenitors fail to exit the cell cycle at the appropriate developmental time and postmitotic cells do not acquire markers of terminal differentiation, and microglial precursors do not colonize the retina. Given the link between Progranulin and cell cycle regulation in peripheral tissues and transformed cells, we analyzed cell cycle kinetics among retinal progenitors following Pgrn-a knockdown. Depleting Pgrn-a results in a significant lengthening of the cell cycle. These data suggest that Pgrn-a plays a dual role during nervous system development by governing the rate at which progenitors progress through the cell cycle and attracting microglial progenitors into the embryonic brain and retina. Collectively, these data show that Pgrn-a governs neurogenesis by regulating cell cycle kinetics and the transition from proliferation to cell cycle exit and differentiation. © 2017 The Authors. Developmental Neurobiology Published by Wiley Periodicals, Inc. Develop Neurobiol 77: 1114-1129, 2017.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Neurogenesis/physiology , Retina/embryology , Retina/metabolism , Zebrafish Proteins/metabolism , Analysis of Variance , Animals , Bromodeoxyuridine/metabolism , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Cyclins/metabolism , Embryo, Nonmammalian , Gene Expression Regulation, Developmental/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Microglia/drug effects , Neurogenesis/drug effects , Neurogenesis/genetics , Oligonucleotides, Antisense/pharmacology , Retina/cytology , Zebrafish , Zebrafish Proteins/genetics
19.
Transl Vis Sci Technol ; 6(2): 5, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28316878

ABSTRACT

This report emerges from a workshop convened by the National Eye Institute (NEI) as part of the "Audacious Goals Initiative" (AGI). The workshop addressed the replacement of retinal ganglion cells (RGCs) from exogenous and endogenous sources, and sought to identify the gaps in our knowledge and barriers to progress in devising cellular replacement therapies for diseases where RGCs die. Here, we briefly review relevant literature regarding common diseases associated with RGC death, the genesis of RGCs in vivo, strategies for generating transplantable RGCs in vitro, and potential endogenous cellular sources to regenerate these cells. These topics provided the clinical and scientific context for the discussion among the workshop participants and are relevant to efforts that may lead to therapeutic approaches for replacing RGCs. This report also summarizes the content of the workshop discussion, which focused on: (1) cell sources for RGC replacement and regeneration, (2) optimizing integration, survival, and synaptogenesis of new RGCs, and (3) approaches for assessing the outcomes of RGC replacement therapies. We conclude this report with a summary of recommendations, based on the workshop discussions, which may guide vision scientists seeking to develop therapies for replacing RGCs in humans.

20.
Invest Ophthalmol Vis Sci ; 56(12): 7496-515, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26580854

ABSTRACT

PURPOSE: Photoreceptor genesis in the retina requires precise regulation of progenitor cell competence, cell cycle exit, and differentiation, although information around the mechanisms that govern these events currently is lacking. In zebrafish, the basic helix-loop-helix (bHLH) transcription factor NeuroD governs photoreceptor genesis, but the signaling pathways through which NeuroD functions are unknown. The purpose of this study was to identify these pathways, and during photoreceptor genesis, Notch signaling was investigated as the putative mediator of NeuroD function. METHODS: In embryos, genetic mosaic analysis was used to determine if NeuroD functions is cell- or non-cell-autonomous. Morpholino-induced NeuroD knockdown, CRISPR/Cas9 mutation, and pharmacologic and transgenic approaches were used, followed by in situ hybridization, immunocytochemistry, and quantitative RT-PCR (qRT-PCR), to identify mechanisms through which NeuroD functions. In adults, following photoreceptor ablation and NeuroD knockdown, similar methods as above were used to identify NeuroD function during photoreceptor regeneration. RESULTS: In embryos, NeuroD function is non-cell-autonomous, NeuroD knockdown increases Notch pathway gene expression, Notch inhibition rescues the NeuroD knockdown-induced deficiency in cell cycle exit but not photoreceptor maturation, and Notch activation and CRISPR/Cas9 mutation of neurod recapitulate NeuroD knockdown. In adults, NeuroD knockdown prevents cell cycle exit and photoreceptor regeneration and increases Notch pathway gene expression, and Notch inhibition rescues this phenotype. CONCLUSIONS: These data demonstrate that during embryonic development, NeuroD governs photoreceptor genesis via non-cell-autonomous mechanisms and that, during photoreceptor development and regeneration, Notch signaling is a mechanistic link between NeuroD and cell cycle exit. In contrast, during embryonic development, NeuroD governs photoreceptor maturation via mechanisms that are independent of Notch signaling.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/genetics , Photoreceptor Cells/metabolism , RNA/genetics , Receptors, Notch/metabolism , Regeneration , Zebrafish/embryology , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cell Differentiation , Cells, Cultured , Helix-Loop-Helix Motifs , Nerve Tissue Proteins/biosynthesis , Photoreceptor Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL