Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Dev Biol ; 433(2): 394-403, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29291983

ABSTRACT

The use of antioxidants in tissue regeneration has been studied, but their mechanism of action is not well understood. Here, we analyze the role of the antioxidant N-acetylcysteine (NAC) in retina regeneration. Embryonic chicks are able to regenerate their retina after its complete removal from retinal stem/progenitor cells present in the ciliary margin (CM) of the eye only if a source of exogenous factors, such as FGF2, is present. This study shows that NAC modifies the redox status of the CM, initiates self-renewal of the stem/progenitor cells, and induces regeneration in the absence of FGF2. NAC works as an antioxidant by scavenging free radicals either independently or through the synthesis of glutathione (GSH), and/or by reducing oxidized proteins through a thiol disulfide exchange activity. We dissected the mechanism used by NAC to induce regeneration through the use of inhibitors of GSH synthesis and the use of other antioxidants with different biochemical structures and modes of action, and found that NAC induces regeneration through its thiol disulfide exchange activity. Thus, our results provide, for the first time, a biochemical basis for induction of retina regeneration. Furthermore, NAC induction was independent of FGF receptor signaling, but dependent on the MAPK (pErk1/2) pathway.


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/pharmacology , Regeneration/drug effects , Retina/physiology , Stem Cells/drug effects , Animals , Cell Differentiation/drug effects , Chick Embryo , Ciliary Body/cytology , Disulfides/metabolism , Fibroblast Growth Factor 2/pharmacology , Glutathione/metabolism , Glutathione Peroxidase/metabolism , MAP Kinase Signaling System/drug effects , Oxidation-Reduction , Regeneration/physiology , Retina/drug effects , Stem Cells/cytology , Sulfhydryl Compounds/metabolism
2.
Dev Biol ; 428(1): 88-100, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28576690

ABSTRACT

Complement components have been implicated in a wide variety of functions including neurogenesis, proliferation, cell migration, differentiation, cancer, and more recently early development and regeneration. Following our initial observations indicating that C3a/C3aR signaling induces chick retina regeneration, we analyzed its role in chick eye morphogenesis. During eye development, the optic vesicle (OV) invaginates to generate a bilayer optic cup (OC) that gives rise to the retinal pigmented epithelium (RPE) and neural retina. We show by immunofluorescence staining that C3 and the receptor for C3a (the cleaved and active form of C3), C3aR, are present in chick embryos during eye morphogenesis in the OV and OC. Interestingly, C3aR is mainly localized in the nuclear compartment at the OC stage. Loss of function studies at the OV stage using morpholinos or a blocking antibody targeting the C3aR (anti-C3aR Ab), causes eye defects such as microphthalmia and defects in the ventral portion of the eye that result in coloboma. Such defects were not observed when C3aR was disrupted at the OC stage. Histological analysis demonstrated that microphthalmic eyes were unable to generate a normal optic stalk or a closed OC. The dorsal/ventral patterning defects were accompanied by an expansion of the ventral markers Pax2, cVax and retinoic acid synthesizing enzyme raldh-3 (aldh1a3) domains, an absence of the dorsal expression of Tbx5 and raldh-1 (aldh1a1) and a re-specification of the ventral RPE to neuroepithelium. In addition, the eyes showed overall decreased expression of Gli1 and a change in distribution of nuclear ß-catenin, suggesting that Shh and Wnt pathways have been affected. Finally, we observed prominent cell death along with a decrease in proliferating cells, indicating that both processes contribute to the microphthalmic phenotype. Together our results show that C3aR is necessary for the proper morphogenesis of the OC. This is the first report implicating C3aR in eye development, revealing an unsuspected hitherto regulator for proper chick eye morphogenesis.


Subject(s)
Body Patterning/physiology , Complement C3a/metabolism , Gene Expression Regulation, Developmental , Receptors, Complement/metabolism , Retinal Pigment Epithelium/embryology , Aldehyde Dehydrogenase/metabolism , Animals , Apoptosis/physiology , Cell Proliferation/physiology , Chick Embryo , Hedgehog Proteins/metabolism , Microphthalmos/embryology , Morphogenesis/physiology , PAX2 Transcription Factor/metabolism , Receptors, Complement/genetics , Retinal Dehydrogenase/metabolism , T-Box Domain Proteins/metabolism , Wnt Signaling Pathway/physiology , Zinc Finger Protein GLI1/biosynthesis , beta Catenin/metabolism
3.
PLoS One ; 9(7): e101748, 2014.
Article in English | MEDLINE | ID: mdl-25003522

ABSTRACT

In the present study we explored the role of ß-catenin in mediating chick retina regeneration. The chick can regenerate its retina by activating stem/progenitor cells present in the ciliary margin (CM) of the eye or via transdifferentiation of the retinal pigmented epithelium (RPE). Both modes require fibroblast growth factor 2 (FGF2). We observed, by immunohistochemistry, dynamic changes of nuclear ß-catenin in the CM and RPE after injury (retinectomy). ß-Catenin nuclear accumulation was transiently lost in cells of the CM in response to injury alone, while the loss of nuclear ß-catenin was maintained as long as FGF2 was present. However, nuclear ß-catenin positive cells remained in the RPE in response to injury and were BrdU-/p27+, suggesting that nuclear ß-catenin prevents those cells from entering the cell cycle. If FGF2 is present, the RPE undergoes dedifferentiation and proliferation concomitant with loss of nuclear ß-catenin. Moreover, retinectomy followed by disruption of active ß-catenin by using a signaling inhibitor (XAV939) or over-expressing a dominant negative form of Lef-1 induces regeneration from both the CM and RPE in the absence of FGF2. Our results imply that ß-catenin protects cells of the CM and RPE from entering the cell cycle in the developing eye, and specifically for the RPE during injury. Thus inactivation of ß-catenin is a pre-requisite for chick retina regeneration.


Subject(s)
Regeneration , Retina/physiology , beta Catenin/metabolism , Animals , Cell Differentiation , Cell Nucleus/metabolism , Cell Proliferation , Chick Embryo , Fibroblast Growth Factor 2/pharmacology , Gene Expression , Gene Expression Regulation, Developmental/drug effects , Heterocyclic Compounds, 3-Ring/pharmacology , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Phenotype , Protein Transport , Retina/cytology , Retina/drug effects , Retina/embryology , Retinal Pigment Epithelium/embryology , Retinal Pigment Epithelium/physiology , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , Transcription, Genetic , Wnt Signaling Pathway/drug effects , beta Catenin/genetics
4.
Nat Commun ; 4: 2312, 2013.
Article in English | MEDLINE | ID: mdl-23942241

ABSTRACT

Identifying the initiation signals for tissue regeneration in vertebrates is one of the major challenges in regenerative biology. Much of the research thus far has indicated that certain growth factors have key roles. Here we show that complement fragment C3a is sufficient to induce complete regeneration of the embryonic chick retina from stem/progenitor cells present in the eye, independent of fibroblast growth factor receptor signaling. Instead, C3a induces retina regeneration via STAT3 activation, which in turn activates the injury- and inflammation-responsive factors, IL-6, IL-8 and TNF-α. This activation sets forth regulation of Wnt2b, Six3 and Sox2, genes associated with retina stem and progenitor cells. Thus, our results establish a mechanism for retina regeneration based on injury and inflammation signals. Furthermore, our results indicate a unique function for complement anaphylatoxins that implicate these molecules in the induction and complete regeneration of the retina, opening new avenues of experimentation in the field.


Subject(s)
Complement C3a/metabolism , Regeneration/physiology , Retina/metabolism , STAT3 Transcription Factor/metabolism , Tissue Engineering/methods , Animals , Chick Embryo , Enzyme Activation , Eye Proteins/metabolism , Guided Tissue Regeneration , Homeodomain Proteins/metabolism , Interleukin-6/biosynthesis , Interleukin-6/metabolism , Interleukin-8/biosynthesis , Interleukin-8/metabolism , MAP Kinase Signaling System , Nerve Tissue Proteins/metabolism , Organ Culture Techniques , Regeneration/immunology , Retina/embryology , Retina/growth & development , SOXB1 Transcription Factors/metabolism , STAT3 Transcription Factor/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/metabolism , Wnt3 Protein/metabolism , Homeobox Protein SIX3
5.
Nat Protoc ; 6(5): 593-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21527917

ABSTRACT

Here we describe a protocol for gene loss of function during regeneration in newts, specifically applied to lens regeneration. Knockdown with the use of morpholinos can be achieved both in vitro and in vivo, depending on the experimental design. These methods achieve desirable levels of gene knockdown, and thus can be compared with methods developed for use in other animals, such as zebrafish. The technology has been applied to study molecular mechanisms during the process of lens regeneration by knocking down genes at specific stages and examining their effects on other genes and lens differentiation. The protocol can take a few days or up to 20 d to complete, depending on the duration of the experiment.


Subject(s)
Gene Knockdown Techniques , Lens, Crystalline/physiology , Salamandridae/genetics , Animals , Models, Animal , Regeneration
6.
Nat Protoc ; 6(5): 600-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21527918

ABSTRACT

The great regenerative abilities of newts provide the impetus for studies at the molecular level. However, efficient methods for gene regulation have historically been quite limited. Here we describe a protocol for transgenically expressing exogenous genes in the newt Cynops pyrrhogaster. This method is simple: a reaction mixture of I-SceI meganuclease and a plasmid DNA carrying a transgene cassette flanked by the enzyme recognition sites is directly injected into fertilized eggs. The protocol achieves a high efficiency of transgenesis, comparable to protocols used in other animal systems, and it provides a practical number of transgenic newts (∼20% of injected embryos) that survive beyond metamorphosis and that can be applied to regenerative studies. The entire protocol for obtaining transgenic adult newts takes 4-5 months.


Subject(s)
Gene Transfer Techniques , Salamandridae/genetics , Animals , Animals, Genetically Modified/genetics , Female , Male
7.
Proc Natl Acad Sci U S A ; 104(51): 20380-5, 2007 Dec 18.
Article in English | MEDLINE | ID: mdl-18093961

ABSTRACT

We identified a mechanism whereby retina regeneration in the embryonic chick can be induced by the contribution of stem/progenitor cells. We show that bone morphogenetic protein (BMP) signaling is sufficient and necessary to induce retina regeneration and that its action can be divided into two phases. By 3 days after postretinectomy (d PR), the BMP pathway directs proliferation and regeneration through the activation of Smad (canonical BMP pathway) and the up-regulation of FGF signaling by the MAPK pathway. By 7d PR, it induces apoptosis by activating p38 (a noncanonical BMP pathway) and down-regulating FGF signaling (by both MAPK and AKT pathways). Apoptosis at this later stage can be prevented, and BMP-induced regeneration can be further induced by inhibition of p38. These results unravel a mechanism for stem/progenitor cell-mediated retina regeneration, where BMP activation establishes a cross-talk with the FGF pathway and selectively activates the canonical and noncanonical BMP pathways. Retina stem/progenitor cells exist in other species, including humans. Thus, our findings provide insights on how retinal stem cells can be activated for possible regenerative therapies.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Fibroblast Growth Factors/metabolism , Regeneration/genetics , Retina/physiology , Stem Cells/physiology , Animals , Apoptosis , Bone Morphogenetic Proteins/genetics , Cell Proliferation , Chick Embryo , Eye/chemistry , Eye/metabolism , Promoter Regions, Genetic , Retina/cytology , Signal Transduction , Smad Proteins/genetics , Stem Cells/cytology , Transcriptional Activation , p38 Mitogen-Activated Protein Kinases/genetics
8.
Proc Natl Acad Sci U S A ; 103(40): 14848-53, 2006 Oct 03.
Article in English | MEDLINE | ID: mdl-17003134

ABSTRACT

Pax-6 is a master regulator of eye development and is expressed in the dorsal and ventral iris during newt lens regeneration. We show that expression of Pax-6 during newt lens regeneration coincides with cell proliferation. By knocking down expression of Pax-6 via treatment with morpholinos, we found that proliferation of iris pigment epithelial cells was dramatically reduced both in vitro and in vivo, and, as a result, lens regeneration was significantly retarded. However, induction of dedifferentiation in the dorsal iris was not inhibited. Pax-6 knockdown early in lens regeneration resulted in inhibition of crystallin expression and retardation of lens fiber induction. Once crystallin expression and differentiation of lens fibers has ensued, however, loss of function of Pax-6 did not affect crystallin expression and lens fiber maintenance, even though the effects on proliferation persisted. These results conclusively show that Pax-6 is associated with distinct early events during lens regeneration, namely control of cell proliferation and subsequent lens fiber differentiation.


Subject(s)
Eye Proteins/metabolism , Homeodomain Proteins/metabolism , Lens, Crystalline/physiology , Paired Box Transcription Factors/metabolism , Regeneration , Repressor Proteins/metabolism , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/genetics , Epithelial Cells/cytology , Iris/cytology , Lens, Crystalline/drug effects , Oligonucleotides, Antisense/pharmacology , PAX6 Transcription Factor , Paired Box Transcription Factors/deficiency , Pigment Epithelium of Eye/cytology , Pigment Epithelium of Eye/drug effects , Regeneration/drug effects , Salamandridae
9.
Curr Neurovasc Res ; 1(3): 231-9, 2004 Jul.
Article in English | MEDLINE | ID: mdl-16181073

ABSTRACT

In this review, we will explore several studies where stem cells from neural, non-neural and even embryonic cells have been used as potential sources to repair the damage retina. In addition, we will also discuss the possibility of inducing retina regeneration by transdifferentiation of cells present in existing eye tissues, such as, the Retinal Pigmented Epithelium (RPE), the Pigmented Ciliary Margin (PCM) and Müller glia cells.


Subject(s)
Retinal Diseases/surgery , Stem Cell Transplantation , Animals , Humans , Physiology/trends , Regeneration , Retina/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...