Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Proc Natl Acad Sci U S A ; 119(15): e2113751119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35394873

ABSTRACT

Although mammalian retinal ganglion cells (RGCs) normally cannot regenerate axons nor survive after optic nerve injury, this failure is partially reversed by inducing sterile inflammation in the eye. Infiltrative myeloid cells express the axogenic protein oncomodulin (Ocm) but additional, as-yet-unidentified, factors are also required. We show here that infiltrative macrophages express stromal cell­derived factor 1 (SDF1, CXCL12), which plays a central role in this regard. Among many growth factors tested in culture, only SDF1 enhances Ocm activity, an effect mediated through intracellular cyclic AMP (cAMP) elevation and phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) activation. SDF1 deficiency in myeloid cells (CXCL12flx/flxLysM-Cre−/+ mice) or deletion of the SDF1 receptor CXCR4 in RGCs (intraocular AAV2-Cre in CXCR4flx/flx mice) or SDF1 antagonist AMD3100 greatly suppresses inflammation-induced regeneration and decreases RGC survival to baseline levels. Conversely, SDF1 induces optic nerve regeneration and RGC survival, and, when combined with Ocm/cAMP, SDF1 increases axon regeneration to levels similar to those induced by intraocular inflammation. In contrast to deletion of phosphatase and tensin homolog (Pten), which promotes regeneration selectively from αRGCs, SDF1 promotes regeneration from non-αRGCs and enables the latter cells to respond robustly to Pten deletion; however, SDF1 surprisingly diminishes the response of αRGCs to Pten deletion. When combined with inflammation and Pten deletion, SDF1 enables many RGCs to regenerate axons the entire length of the optic nerve. Thus, SDF1 complements the effects of Ocm in mediating inflammation-induced regeneration and enables different RGC subtypes to respond to Pten deletion.


Subject(s)
Optic Nerve Injuries , Retinal Ganglion Cells , Axons/metabolism , Chemokine CXCL12/genetics , Monocytes/metabolism , Nerve Regeneration/physiology , Optic Nerve Injuries/genetics , Optic Nerve Injuries/metabolism , PTEN Phosphohydrolase/genetics , Retinal Ganglion Cells/physiology
2.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Article in English | MEDLINE | ID: mdl-33627402

ABSTRACT

Ciliary neurotrophic factor (CNTF) is a leading therapeutic candidate for several ocular diseases and induces optic nerve regeneration in animal models. Paradoxically, however, although CNTF gene therapy promotes extensive regeneration, recombinant CNTF (rCNTF) has little effect. Because intraocular viral vectors induce inflammation, and because CNTF is an immune modulator, we investigated whether CNTF gene therapy acts indirectly through other immune mediators. The beneficial effects of CNTF gene therapy remained unchanged after deleting CNTF receptor alpha (CNTFRα) in retinal ganglion cells (RGCs), the projection neurons of the retina, but were diminished by depleting neutrophils or by genetically suppressing monocyte infiltration. CNTF gene therapy increased expression of C-C motif chemokine ligand 5 (CCL5) in immune cells and retinal glia, and recombinant CCL5 induced extensive axon regeneration. Conversely, CRISPR-mediated knockdown of the cognate receptor (CCR5) in RGCs or treating wild-type mice with a CCR5 antagonist repressed the effects of CNTF gene therapy. Thus, CCL5 is a previously unrecognized, potent activator of optic nerve regeneration and mediates many of the effects of CNTF gene therapy.


Subject(s)
Chemokine CCL5/metabolism , Ciliary Neurotrophic Factor/genetics , Nerve Regeneration , Optic Nerve/metabolism , Animals , CRISPR-Cas Systems , Chemokine CCL5/genetics , Ciliary Neurotrophic Factor/metabolism , Ciliary Neurotrophic Factor Receptor alpha Subunit/genetics , Ciliary Neurotrophic Factor Receptor alpha Subunit/metabolism , Gene Editing , Genetic Therapy , Genetic Vectors/genetics , Nerve Regeneration/genetics , Optic Nerve Injuries/etiology , Optic Nerve Injuries/therapy , Retinal Ganglion Cells/metabolism
3.
Int J Mol Sci ; 24(20)2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37895039

ABSTRACT

Although most pathways in the mature central nervous system cannot regenerate when injured, research beginning in the late 20th century has led to discoveries that may help reverse this situation. Here, we highlight research in recent years from our laboratory identifying oncomodulin (Ocm), stromal cell-derived factor (SDF)-1, and chemokine CCL5 as growth factors expressed by cells of the innate immune system that promote axon regeneration in the injured optic nerve and elsewhere in the central and peripheral nervous systems. We also review the role of ArmC10, a newly discovered Ocm receptor, in mediating many of these effects, and the synergy between inflammation-derived growth factors and complementary strategies to promote regeneration, including deleting genes encoding cell-intrinsic suppressors of axon growth, manipulating transcription factors that suppress or promote the expression of growth-related genes, and manipulating cell-extrinsic suppressors of axon growth. In some cases, combinatorial strategies have led to unprecedented levels of nerve regeneration. The identification of some similar mechanisms in human neurons offers hope that key discoveries made in animal models may eventually lead to treatments to improve outcomes after neurological damage in patients.


Subject(s)
Axons , Nerve Regeneration , Animals , Humans , Axons/physiology , Nerve Regeneration/physiology , Optic Nerve/physiology , Neurons , Central Nervous System , Intercellular Signaling Peptides and Proteins
4.
Proc Natl Acad Sci U S A ; 114(2): E209-E218, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28049831

ABSTRACT

Retinal ganglion cells (RGCs), the projection neurons of the eye, cannot regenerate their axons once the optic nerve has been injured and soon begin to die. Whereas RGC death and regenerative failure are widely viewed as being cell-autonomous or influenced by various types of glia, we report here that the dysregulation of mobile zinc (Zn2+) in retinal interneurons is a primary factor. Within an hour after the optic nerve is injured, Zn2+ increases several-fold in retinal amacrine cell processes and continues to rise over the first day, then transfers slowly to RGCs via vesicular release. Zn2+ accumulation in amacrine cell processes involves the Zn2+ transporter protein ZnT-3, and deletion of slc30a3, the gene encoding ZnT-3, promotes RGC survival and axon regeneration. Intravitreal injection of Zn2+ chelators enables many RGCs to survive for months after nerve injury and regenerate axons, and enhances the prosurvival and regenerative effects of deleting the gene for phosphatase and tensin homolog (pten). Importantly, the therapeutic window for Zn2+ chelation extends for several days after nerve injury. These results show that retinal Zn2+ dysregulation is a major factor limiting the survival and regenerative capacity of injured RGCs, and point to Zn2+ chelation as a strategy to promote long-term RGC protection and enhance axon regeneration.


Subject(s)
Nerve Regeneration , Optic Nerve Injuries/metabolism , Optic Nerve/physiology , Retina/physiology , Zinc/metabolism , Animals , Carrier Proteins/genetics , Carrier Proteins/physiology , Cation Transport Proteins , Chelating Agents/pharmacology , Ethylamines/pharmacology , Male , Membrane Proteins/genetics , Membrane Proteins/physiology , Membrane Transport Proteins , Mice, Inbred C57BL , Mice, Knockout , Pyridines/pharmacology , Sulfanilic Acids/pharmacology
5.
Proc Natl Acad Sci U S A ; 109(23): 9149-54, 2012 Jun 05.
Article in English | MEDLINE | ID: mdl-22615390

ABSTRACT

The mature optic nerve cannot regenerate when injured, leaving victims of traumatic nerve damage or diseases such as glaucoma with irreversible visual losses. Recent studies have identified ways to stimulate retinal ganglion cells to regenerate axons part-way through the optic nerve, but it remains unknown whether mature axons can reenter the brain, navigate to appropriate target areas, or restore vision. We show here that with adequate stimulation, retinal ganglion cells are able to regenerate axons the full length of the visual pathway and on into the lateral geniculate nucleus, superior colliculus, and other visual centers. Regeneration partially restores the optomotor response, depth perception, and circadian photoentrainment, demonstrating the feasibility of reconstructing central circuitry for vision after optic nerve damage in mature mammals.


Subject(s)
Axons/physiology , Optic Nerve Injuries/drug therapy , Optic Nerve/physiology , Regeneration/physiology , Retinal Ganglion Cells/physiology , Zymosan/pharmacology , Animals , Calcium-Binding Proteins/metabolism , Circadian Rhythm/physiology , Cyclic AMP/metabolism , Dependovirus , Gene Deletion , Genetic Vectors/genetics , Geniculate Bodies/physiology , Integrases/metabolism , Mice , Optic Nerve/cytology , PTEN Phosphohydrolase/genetics , Retinal Ganglion Cells/cytology , Superior Colliculi/physiology , Zymosan/administration & dosage
6.
J Neurosci ; 33(37): 14816-24, 2013 Sep 11.
Article in English | MEDLINE | ID: mdl-24027282

ABSTRACT

Although neurons are normally unable to regenerate their axons after injury to the CNS, this situation can be partially reversed by activating the innate immune system. In a widely studied instance of this phenomenon, proinflammatory agents have been shown to cause retinal ganglion cells, the projection neurons of the eye, to regenerate lengthy axons through the injured optic nerve. However, the role of different molecules and cell populations in mediating this phenomenon remains unclear. We show here that neutrophils, the first responders of the innate immune system, play a central role in inflammation-induced regeneration. Numerous neutrophils enter the mouse eye within a few hours of inducing an inflammatory reaction and express high levels of the atypical growth factor oncomodulin (Ocm). Immunodepletion of neutrophils diminished Ocm levels in the eye without altering levels of CNTF, leukemia inhibitory factor, or IL-6, and suppressed the proregenerative effects of inflammation. A peptide antagonist of Ocm suppressed regeneration as effectively as neutrophil depletion. Macrophages enter the eye later in the inflammatory process but appear to be insufficient to stimulate extensive regeneration in the absence of neutrophils. These data provide the first evidence that neutrophils are a major source of Ocm and can promote axon regeneration in the CNS.


Subject(s)
Calcium-Binding Proteins/metabolism , Nerve Regeneration/physiology , Neutrophils/metabolism , Optic Nerve Diseases/pathology , Optic Nerve Diseases/physiopathology , Animals , Antigens, CD/metabolism , Cells, Cultured , Ciliary Neurotrophic Factor/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-6/metabolism , Leukemia Inhibitory Factor/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Crush , Receptors, Cell Surface/metabolism , Retina/cytology , Retinal Ganglion Cells/metabolism , Stilbamidines/metabolism , Visual Pathways/physiology
7.
Sci Transl Med ; 15(708): eadg6241, 2023 08 09.
Article in English | MEDLINE | ID: mdl-37556559

ABSTRACT

Oncomodulin (Ocm) is a myeloid cell-derived growth factor that enables axon regeneration in mice and rats after optic nerve injury or peripheral nerve injury, yet the mechanisms underlying its activity are unknown. Using proximity biotinylation, coimmunoprecipitation, surface plasmon resonance, and ectopic expression, we have identified armadillo-repeat protein C10 (ArmC10) as a high-affinity receptor for Ocm. ArmC10 deletion suppressed inflammation-induced axon regeneration in the injured optic nerves of mice. ArmC10 deletion also suppressed the ability of lesioned sensory neurons to regenerate peripheral axons rapidly after a second injury and to regenerate their central axons after spinal cord injury in mice (the conditioning lesion effect). Conversely, Ocm acted through ArmC10 to accelerate optic nerve and peripheral nerve regeneration and to enable spinal cord axon regeneration in these mouse nerve injury models. We showed that ArmC10 is highly expressed in human-induced pluripotent stem cell-derived sensory neurons and that exposure to Ocm altered gene expression and enhanced neurite outgrowth. ArmC10 was also expressed in human monocytes, and Ocm increased the expression of immune modulatory genes in these cells. These findings suggest that Ocm acting through its receptor ArmC10 may be a useful therapeutic target for nerve repair and immune modulation.


Subject(s)
Axons , Induced Pluripotent Stem Cells , Animals , Humans , Mice , Ganglia, Spinal/metabolism , Nerve Regeneration , Neuronal Outgrowth , Sensory Receptor Cells
8.
Proc Natl Acad Sci U S A ; 106(46): 19587-92, 2009 Nov 17.
Article in English | MEDLINE | ID: mdl-19875691

ABSTRACT

The inflammatory response that accompanies central nervous system (CNS) injury can affect neurological outcome in both positive and negative ways. In the optic nerve, a CNS pathway that normally fails to regenerate when damaged, intraocular inflammation causes retinal ganglion cells (RGCs) to switch into an active growth state and extend lengthy axons down the nerve. The molecular basis of this phenomenon is uncertain. A prior study showed that oncomodulin (Ocm), a Ca(2+)-binding protein secreted by a macrophage cell line, is a potent axon-promoting factor for RGCs. However, it is not known whether Ocm contributes to the physiological effects of intraocular inflammation in vivo, and there are conflicting reports in the literature regarding its expression and significance. We show here that intraocular inflammation causes infiltrative cells of the innate immune system to secrete high levels of Ocm, and that agents that prevent Ocm from binding to its receptor suppress axon regeneration. These results were verified in different strains, species, and experimental models, and establish Ocm as a potent growth-promoting signal between the innate immune system and neurons in vivo.


Subject(s)
Axons/physiology , Calcium-Binding Proteins/physiology , Inflammation/immunology , Nerve Regeneration , Optic Nerve/physiology , Animals , Axons/metabolism , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/metabolism , Cell Line , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley
9.
Genes (Basel) ; 13(6)2022 05 31.
Article in English | MEDLINE | ID: mdl-35741750

ABSTRACT

Non-heading Chinese cabbage (Brassica campestris ssp. chinensis) is an important vegetative crop in the south of China. As an antioxidant, anthocyanin is the major quality trait for vegetables with purple leaves or petioles. However, the molecular biosynthetic mechanism of anthocyanin in non-heading Chinese cabbage has not been explained exclusively. In this study, two non-heading Chinese cabbage with contrasting colors in the leaves were used as the materials for RNA-seq. A total of 906 DEGs were detected, and we found that the anthocyanin and flavonoid biosynthetic pathways are significantly enriched in the purple NHCC. The transcriptome result was verified by RT-qPCR. Though bioinformatics analysis, BcTT8 was selected as the candidate gene for the regulation of anthocyanin synthesis, and the characterization of BcTT8 was elucidated by the functional analyses. The results proved that BcTT8 is a nucleus protein and phylogenetically close to the TT8 protein from Brassica. After silencing BcTT8, the total anthocyanin content of pTY-BcTT8 plants decreased by 42.5%, and the relative expression levels of anthocyanin pathway genes BcDFR, BcLODX and BcUF3GT-1 were significantly downregulated, while the transcription level of BcFLS was significantly upregulated. Compared with the wild type, the transgenic Arabidopsis showed obvious violet in the cotyledons part, and the anthocyanin biosynthetic genes such as AtDFR and AtLODX were significantly upregulated. In conclusion, BcTT8 is critical in the anthocyanin synthesis process of non-heading Chinese cabbage. Our findings illustrated the molecular mechanism of anthocyanin biosynthesis in non-heading Chinese cabbage.


Subject(s)
Brassica , Transcriptome , Anthocyanins , Brassica/genetics , Brassica/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Plant , Transcriptome/genetics
10.
Sci Rep ; 12(1): 17446, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36261683

ABSTRACT

Adult central nervous system (CNS) axons fail to regenerate after injury, and master regulators of the regenerative program remain to be identified. We analyzed the transcriptomes of retinal ganglion cells (RGCs) at 1 and 5 days after optic nerve injury with and without a cocktail of strongly pro-regenerative factors to discover genes that regulate survival and regeneration. We used advanced bioinformatic analysis to identify the top transcriptional regulators of upstream genes and cross-referenced these with the regulators upstream of genes differentially expressed between embryonic RGCs that exhibit robust axon growth vs. postnatal RGCs where this potential has been lost. We established the transcriptional activator Elk-1 as the top regulator of RGC gene expression associated with axon outgrowth in both models. We demonstrate that Elk-1 is necessary and sufficient to promote RGC neuroprotection and regeneration in vivo, and is enhanced by manipulating specific phosphorylation sites. Finally, we co-manipulated Elk-1, PTEN, and REST, another transcription factor discovered in our analysis, and found Elk-1 to be downstream of PTEN and inhibited by REST in the survival and axon regenerative pathway in RGCs. These results uncover the basic mechanisms of regulation of survival and axon growth and reveal a novel, potent therapeutic strategy to promote neuroprotection and regeneration in the adult CNS.


Subject(s)
Optic Nerve Injuries , Retinal Ganglion Cells , Humans , Retinal Ganglion Cells/metabolism , Axons/metabolism , Nerve Regeneration/physiology , Optic Nerve Injuries/genetics , Optic Nerve Injuries/metabolism , Transcription Factors/metabolism
11.
Nat Commun ; 13(1): 4418, 2022 07 29.
Article in English | MEDLINE | ID: mdl-35906210

ABSTRACT

The inability of neurons to regenerate long axons within the CNS is a major impediment to improving outcome after spinal cord injury, stroke, and other CNS insults. Recent advances have uncovered an intrinsic program that involves coordinate regulation by multiple transcription factors that can be manipulated to enhance growth in the peripheral nervous system. Here, we use a systems genomics approach to characterize regulatory relationships of regeneration-associated transcription factors, identifying RE1-Silencing Transcription Factor (REST; Neuron-Restrictive Silencer Factor, NRSF) as a predicted upstream suppressor of a pro-regenerative gene program associated with axon regeneration in the CNS. We validate our predictions using multiple paradigms, showing that mature mice bearing cell type-specific deletions of REST or expressing dominant-negative mutant REST show improved regeneration of the corticospinal tract and optic nerve after spinal cord injury and optic nerve crush, which is accompanied by upregulation of regeneration-associated genes in cortical motor neurons and retinal ganglion cells, respectively. These analyses identify a role for REST as an upstream suppressor of the intrinsic regenerative program in the CNS and demonstrate the utility of a systems biology approach involving integrative genomics and bio-informatics to prioritize hypotheses relevant to CNS repair.


Subject(s)
Axons , Repressor Proteins/metabolism , Spinal Cord Injuries , Animals , Axons/physiology , Mice , Nerve Regeneration/genetics , Retinal Ganglion Cells/physiology , Spinal Cord Injuries/genetics , Spinal Cord Injuries/therapy , Transcription Factors/genetics
12.
J Neurosci ; 30(46): 15654-63, 2010 Nov 17.
Article in English | MEDLINE | ID: mdl-21084621

ABSTRACT

The inability of retinal ganglion cells (RGCs) to regenerate damaged axons through the optic nerve has dire consequences for victims of traumatic nerve injury and certain neurodegenerative diseases. Several strategies have been shown to induce appreciable regeneration in vivo, but the regrowth of axons through the entire optic nerve and on into the brain remains a major challenge. We show here that the induction of a controlled inflammatory response in the eye, when combined with elevation of intracellular cAMP and deletion of the gene encoding pten (phosphatase and tensin homolog), enables RGCs to regenerate axons the full length of the optic nerve in mature mice; approximately half of these axons cross the chiasm, and a rare subset (∼1%) manages to enter the thalamus. Consistent with our previous findings, the axon-promoting effects of inflammation were shown to require the macrophage-derived growth factor Oncomodulin (Ocm). Elevation of cAMP increased the ability of Ocm to bind to its receptors in the inner retina and augmented inflammation-induced regeneration twofold. Inflammation combined with elevated cAMP and PTEN deletion increased activation of the phosphatidylinositol 3-kinase and mitogen-activated protein kinase signaling pathways and augmented regeneration ∼10-fold over the level induced by either pten deletion or Zymosan alone. Thus, treatments that synergistically alter the intrinsic growth state of RGCs produce unprecedented levels of axon regeneration in the optic nerve, a CNS pathway long believed to be incapable of supporting such growth.


Subject(s)
Axons/physiology , Calcium-Binding Proteins/physiology , Cyclic AMP/physiology , Nerve Regeneration/physiology , Optic Nerve/physiology , PTEN Phosphohydrolase/deficiency , Animals , Gene Deletion , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Regeneration/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/physiology , Rats , Rats, Inbred F344
13.
Nat Neurosci ; 9(6): 843-52, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16699509

ABSTRACT

The optic nerve, like most mature CNS pathways, does not regenerate after injury. Through unknown mechanisms, however, macrophage activation in the eye stimulates retinal ganglion cells (RGCs) to regenerate long axons beyond the site of optic nerve injury. Here we identify the calcium (Ca(2+))-binding protein oncomodulin as a potent macrophage-derived growth factor for RGCs and other neurons. Oncomodulin binds to rat RGCs with high affinity in a cyclic AMP (cAMP)-dependent manner and stimulates more extensive outgrowth than other known trophic agents. Depletion of oncomodulin from macrophage-conditioned media (MCM) eliminates the axon-promoting activity of MCM. The effects of oncomodulin involve downstream signaling via Ca(2+)/calmodulin kinase and gene transcription. In vivo, oncomodulin released from microspheres promotes regeneration in the mature rat optic nerve. Oncomodulin also stimulates outgrowth from peripheral sensory neurons. Thus, oncomodulin is a new growth factor for neurons of the mature central and peripheral nervous systems.


Subject(s)
Calcium-Binding Proteins/physiology , Growth Cones/metabolism , Macrophages/metabolism , Nerve Regeneration/physiology , Retinal Ganglion Cells/metabolism , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/physiology , Calcium-Binding Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 1 , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Communication/drug effects , Cell Communication/physiology , Cells, Cultured , Culture Media, Conditioned/pharmacology , Cyclic AMP/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Growth Cones/drug effects , Growth Cones/ultrastructure , Growth Substances/metabolism , Male , Nerve Growth Factors/metabolism , Nerve Regeneration/drug effects , Optic Nerve/cytology , Optic Nerve/drug effects , Optic Nerve/metabolism , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Transcriptional Activation/physiology
14.
iScience ; 23(2): 100836, 2020 Feb 21.
Article in English | MEDLINE | ID: mdl-32058951

ABSTRACT

Growth cones (GCs) are structures associated with growing neurons. GC membrane expansion, which necessitates protein-lipid interactions, is critical to axonal elongation in development and in adult neuritogenesis. We present a multi-omic analysis that integrates proteomics and lipidomics data for the identification of GC pathways, cell phenotypes, and lipid-protein interactions, with an analytic platform to facilitate the visualization of these data. We combine lipidomic data from GC and adult axonal regeneration following optic nerve crush. Our results reveal significant molecular variability in GCs across developmental ages that aligns with the upregulation and downregulation of lipid metabolic processes and correlates with distinct changes in the lipid composition of GC plasmalemma. We find that these processes also define the transition into a growth-permissive state in the adult central nervous system. The insight derived from these analyses will aid in promoting adult regeneration and functional innervation in devastating neurodegenerative diseases.

15.
Restor Neurol Neurosci ; 37(6): 525-544, 2019.
Article in English | MEDLINE | ID: mdl-31609715

ABSTRACT

The optic nerve conveys information about the outside world from the retina to multiple subcortical relay centers. Until recently, the optic nerve was widely believed to be incapable of re-growing if injured, with dire consequences for victims of traumatic, ischemic, or neurodegenerative diseases of this pathway. Over the past 10-20 years, research from our lab and others has made considerable progress in defining factors that normally suppress axon regeneration and the ability of retinal ganglion cells, the projection neurons of the retina, to survive after nerve injury. Here we describe research from our lab on the role of inflammation-derived growth factors, suppression of inter-cellular signals among diverse retinal cell types, and combinatorial therapies, along with related studies from other labs, that enable animals with optic nerve injury to regenerate damaged retinal axons back to the brain. These studies raise the possibility that vision might one day be restored to people with optic nerve damage.


Subject(s)
Nerve Regeneration/physiology , Optic Nerve Injuries/metabolism , Optic Nerve/physiology , Retinal Ganglion Cells/metabolism , Animals , Axons/metabolism , Axons/ultrastructure , Humans , Inflammation Mediators/metabolism , Optic Nerve/ultrastructure , Optic Nerve Injuries/pathology , Retinal Ganglion Cells/ultrastructure
16.
Methods Mol Biol ; 1695: 187-205, 2018.
Article in English | MEDLINE | ID: mdl-29190028

ABSTRACT

Glaucoma is marked by a progressive degeneration of the optic nerve and delayed loss of retinal ganglion cells (RGCs), the projection neurons of the eye. Because RGCs are not replaced and because surviving RGCs cannot regenerate their axons, the visual loss in glaucoma is largely irreversible. Here, we describe methods to evaluate treatments that may be beneficial for treating glaucoma using in vitro cell culture models (immunopanning to isolate neonatal RGCs, dissociated mature retinal neurons, retinal explants) and in vivo models that test potential treatments or investigate underlying molecular mechanisms in an intact system. Potentially, use of these models can help investigators continue to improve treatments to preserve RGCs and restore visual function in patients with glaucoma.


Subject(s)
Glaucoma/therapy , Nerve Regeneration , Retinal Ganglion Cells/cytology , Animals , Cell Culture Techniques , Cell Survival , Cells, Cultured , In Vitro Techniques , Mice , Rats , Retinal Ganglion Cells/physiology
17.
J Neurosci ; 23(21): 7830-8, 2003 Aug 27.
Article in English | MEDLINE | ID: mdl-12944512

ABSTRACT

Mammalian retinal ganglion cells (RGCs) do not normally regenerate their axons through an injured optic nerve, but can be stimulated to do so by activating macrophages intraocularly. In a cell culture model of this phenomenon, we found that a small molecule that is constitutively present in the vitreous, acting in concert with macrophage-derived proteins, stimulates mature rat RGCs to regenerate their axons if intracellular cAMP is elevated. In lower vertebrates, RGCs regenerate their axons spontaneously in vivo, and in culture, the most potent axon-promoting factor for these cells is a molecule that resembles the small vitreous-derived growth factor from the rat. This molecule was isolated chromatographically and was shown by mass spectrometry to be a carbohydrate. In agreement with this finding, D-mannose proved to be a potent axon-promoting factor for rat RGCs (ED50 approximately 10 microm); this response was cAMP-dependent and was augmented further by macrophage-derived proteins. Goldfish RGCs showed far less selectivity, responding strongly to either D-mannose or D-glucose in a cAMP-independent manner. These findings accord well with the success or failure of optic nerves to regenerate in higher and lower vertebrates in vivo. The axon-promoting effects of mannose are highly specific and are unrelated to energy metabolism or glycoprotein synthesis.


Subject(s)
Axons/physiology , Carbohydrates/pharmacology , Cyclic AMP/metabolism , Nerve Growth Factors/pharmacology , Nerve Regeneration , Retinal Ganglion Cells/physiology , Animals , Axons/drug effects , Carbohydrates/chemistry , Carbohydrates/isolation & purification , Cells, Cultured , Goldfish , Macrophages/chemistry , Male , Mannose/pharmacology , Mass Spectrometry , Nerve Growth Factors/chemistry , Nerve Growth Factors/isolation & purification , Neuroglia/physiology , Rats , Rats, Inbred F344 , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/drug effects , Vitreous Body/cytology
18.
J Neurosci ; 23(6): 2284-93, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12657687

ABSTRACT

After optic nerve injury in mature mammals, retinal ganglion cells (RGCs) are normally unable to regenerate their axons and undergo delayed apoptosis. However, if the lens is damaged at the time of nerve injury, many RGCs survive axotomy and regenerate their axons into the distal optic nerve. Lens injury induces macrophage activation, and we show here that factors secreted by macrophages stimulate RGCs to regenerate their axons. When macrophages were activated by intravitreal injections of Zymosan, a yeast cell wall preparation, the number of RGC axons regenerating into the distal optic nerve was even greater than after lens injury. These effects were further enhanced if Zymosan was injected 3 d after nerve crush. In a grafting paradigm, intravitreal Zymosan increased the number of RGCs that regenerated their axons through a 1.5 cm peripheral nerve graft twofold relative to uninjected controls and threefold if injections were delayed 3 d. In cell culture, media conditioned by activated macrophages stimulated adult rat RGCs to regenerate their axons; this effect was potentiated by a low molecular weight factor that is constitutively present in the vitreous humor. After gel-filtration chromatography, macrophage-derived proteins > or =30 kDa were found to be toxic to RGCs, whereas proteins <30 kDa reversed this toxicity and promoted axon regeneration. The protein(s) that stimulated axon growth is distinct from identified polypeptide trophic factors that were tested. Thus, macrophages produce proteins with both positive and negative effects on RGCs, and the effects of macrophages can be optimized by the timing of their activation.


Subject(s)
Growth Substances/pharmacology , Macrophages/metabolism , Nerve Regeneration/physiology , Optic Nerve/physiology , Animals , Axons/physiology , Cell Count , Culture Media, Conditioned/chemistry , Culture Media, Conditioned/pharmacology , Fluorescent Dyes , GAP-43 Protein/biosynthesis , Growth Substances/metabolism , Immunohistochemistry , Macrophage Activation/drug effects , Macrophages/drug effects , Male , Molecular Weight , Nerve Crush , Nerve Regeneration/drug effects , Optic Nerve/surgery , Peripheral Nerves/transplantation , Rats , Rats, Inbred F344 , Receptors, Interferon/physiology , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/physiology , Sciatic Nerve/transplantation , Time Factors , Zymosan/pharmacology
19.
Nat Neurosci ; 18(12): 1737-45, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26502261

ABSTRACT

Stroke produces a limited process of neural repair. Axonal sprouting in cortex adjacent to the infarct is part of this recovery process, but the signal that initiates axonal sprouting is not known. Growth and differentiation factor 10 (GDF10) is induced in peri-infarct neurons in mice, non-human primates and humans. GDF10 promotes axonal outgrowth in vitro in mouse, rat and human neurons through TGFßRI and TGFßRII signaling. Using pharmacogenetic gain- and loss-of-function studies, we found that GDF10 produced axonal sprouting and enhanced functional recovery after stroke; knocking down GDF10 blocked axonal sprouting and reduced recovery. RNA sequencing from peri-infarct cortical neurons revealed that GDF10 downregulated PTEN, upregulated PI3 kinase signaling and induced specific axonal guidance molecules. Using unsupervised genome-wide association analysis of the GDF10 transcriptome, we found that it was not related to neurodevelopment, but may partially overlap with other CNS injury patterns. Thus, GDF10 is a stroke-induced signal for axonal sprouting and functional recovery.


Subject(s)
Axons/metabolism , Growth Differentiation Factor 10/biosynthesis , Growth Differentiation Factor 10/genetics , Recovery of Function/physiology , Stroke/genetics , Stroke/metabolism , Aged , Aged, 80 and over , Animals , Animals, Newborn , Axons/pathology , Humans , Macaca mulatta , Mice , Mice, Inbred C57BL , Middle Aged , Retrospective Studies , Stroke/pathology
20.
Neuron ; 86(5): 1215-27, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-26004914

ABSTRACT

Axon regeneration in the CNS requires reactivating injured neurons' intrinsic growth state and enabling growth in an inhibitory environment. Using an inbred mouse neuronal phenotypic screen, we find that CAST/Ei mouse adult dorsal root ganglion neurons extend axons more on CNS myelin than the other eight strains tested, especially when pre-injured. Injury-primed CAST/Ei neurons also regenerate markedly in the spinal cord and optic nerve more than those from C57BL/6 mice and show greater sprouting following ischemic stroke. Heritability estimates indicate that extended growth in CAST/Ei neurons on myelin is genetically determined, and two whole-genome expression screens yield the Activin transcript Inhba as most correlated with this ability. Inhibition of Activin signaling in CAST/Ei mice diminishes their CNS regenerative capacity, whereas its activation in C57BL/6 animals boosts regeneration. This screen demonstrates that mammalian CNS regeneration can occur and reveals a molecular pathway that contributes to this ability.


Subject(s)
Axons/physiology , Ganglia, Spinal/physiology , Nerve Regeneration/physiology , Sciatic Neuropathy/physiopathology , Spinal Cord Injuries/physiopathology , Animals , Mice , Mice, 129 Strain , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred NOD , Sciatic Neuropathy/pathology , Spinal Cord Injuries/pathology
SELECTION OF CITATIONS
SEARCH DETAIL