Larry I. Benowitz’s research while affiliated with Boston Children's Hospital and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (198)


Disruption of G3BP1 granules promotes mammalian CNS and PNS axon regeneration
  • Article

February 2025

·

45 Reads

Proceedings of the National Academy of Sciences

·

Manasi Agrawal

·

Nicholas Hanovice

·

[...]

·

Jeffery L Twiss

Depletion or inhibition of core stress granule proteins, G3BP1 in mammals and TIAR-2 in Caenorhabditis elegans , increases the growth of spontaneously regenerating axons. Inhibition of G3BP1 by expression of its acidic or “B-domain” accelerates axon regeneration after nerve injury, bringing a potential therapeutic strategy for peripheral nerve repair. Here, we asked whether G3BP1 inhibition is a viable strategy to promote regeneration in injured mammalian central nervous system (CNS) where axons do not regenerate spontaneously. G3BP1 B-domain expression was found to promote axon regeneration in the transected spinal cord provided with a permissive peripheral nerve graft (PNG) as well as in crushed optic nerve. Moreover, a cell-permeable peptide (CPP) to a subregion of B-domain (rodent G3BP1 amino acids 190 to 208) accelerated axon regeneration after peripheral nerve injury and promoted regrowth of reticulospinal axons into the distal transected spinal cord through a bridging PNG. G3BP1 CPP promoted axon growth from rodent and human neurons cultured on permissive substrates, and this function required alternating Glu/Asp-Pro repeats that impart a unique predicted tertiary structure. The G3BP1 CPP disassembles axonal G3BP1, G3BP2, and FMRP, but not FXR1, granules and selectively increases axonal protein synthesis in cortical neurons. These studies identify G3BP1 granules as a key regulator of axon growth in CNS neurons and demonstrate that disassembly of these granules promotes retinal axon regeneration in injured optic nerve and reticulospinal axon elongation into permissive environments after CNS injury. This work highlights G3BP1 granule disassembly as a potential therapeutic strategy for enhancing axon growth and neural repair.


In Vitro and In Vivo Methods for Studying Retinal Ganglion Cell Survival and Optic Nerve Regeneration

October 2024

·

7 Reads

Methods in molecular biology (Clifton, N.J.)

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, the use of these models can help investigators continue to improve treatments to preserve RGCs and restore visual function in patients with glaucoma.


Disruption of Core Stress Granule Protein Aggregates Promotes CNS Axon Regeneration

June 2024

·

42 Reads

Depletion or inhibition of core stress granule proteins, G3BP1 in mammals and TIAR-2 in C. elegans , increases axon regeneration in injured neurons, showing spontaneous regeneration. Inhibition of G3BP1 by expression of its acidic or ‘B-domain’ accelerates axon regeneration after nerve injury, bringing a potential therapeutic intervention to promote neural repair in the peripheral nervous system. Here, we asked if G3BP1 inhibition is a viable strategy to promote regeneration in injured mammalian central nervous system where axons do not regenerate spontaneously. G3BP1 B-domain expression was found to promote axon regeneration in the transected spinal cord provided with a permissive peripheral nerve graft (PNG) as well as in crushed optic nerve. Moreover, a cell-permeable peptide (CPP) to a subregion of B-domain (rodent G3BP1 amino acids 190-208) accelerated axon regeneration after peripheral nerve injury and promoted regrowth of reticulospinal axons into the distal transected spinal cord through a bridging PNG. G3BP1 CPP promoted axon growth from rodent and human neurons cultured on permissive substrates, and this function required alternating Glu/Asp-Pro repeats that impart a unique predicted tertiary structure. The G3BP1 CPP disassembles axonal G3BP1, G3BP2, and FMRP, but not FXR1, granules and selectively increases axonal protein synthesis in cortical neurons. These studies identify G3BP1 granules as a key regulator of axon growth in CNS neurons and demonstrate that disassembly of these granules promotes retinal axon regeneration in injured optic nerve and reticulospinal axon elongation into permissive environments after CNS injury. This work highlights G3BP1 granule disassembly as a potential therapeutic strategy for enhancing axon growth and neural repair. SIGNIFICANCE STATEMENT The central nervous system (CNS) axon does not have the capacity for spontaneous axon regeneration, as seen in the peripheral nervous system (PNS). We previously showed that stress granule-like aggregates of G3BP1 are present in uninjured PNS axons, and these slow nerve regeneration. We now report that CNS axons contain G3BP1 granules, and G3BP1 granule disassembling strategies promote axon regeneration in the injured sciatic nerve, transected spinal cord with a peripheral nerve graft, and injured optic nerve. Thus, G3BP1 granules are a barrier to axon regeneration and can be targeted for stimulating neural repair following traumatic injury, including in the regeneration refractory CNS.


Ca 2+ /Calmodulin-Dependent Protein Kinase II Enhances Retinal Ganglion Cell Survival But Suppresses Axon Regeneration after Optic Nerve Injury
  • Article
  • Full-text available

March 2024

·

55 Reads

·

6 Citations

eNeuro

Neuroprotection after injury or in neurodegenerative disease remains a major goal for basic and translational neuroscience. Retinal ganglion cells (RGCs), the projection neurons of the eye, degenerate in optic neuropathies after axon injury, and there are no clinical therapies to prevent their loss or restore their connectivity to targets in the brain. Here we demonstrate a profound neuroprotective effect of the exogenous expression of various Ca ²⁺ /calmodulin-dependent protein kinase II (CaMKII) isoforms in mice. A dramatic increase in RGC survival following the optic nerve trauma was elicited by the expression of constitutively active variants of multiple CaMKII isoforms in RGCs using adeno-associated viral (AAV) vectors across a 100-fold range of AAV dosing in vivo. Despite this neuroprotection, however, short-distance RGC axon sprouting was suppressed by CaMKII, and long-distance axon regeneration elicited by several pro-axon growth treatments was likewise inhibited even as CaMKII further enhanced RGC survival. Notably, in a dose-escalation study, AAV-expressed CaMKII was more potent for axon growth suppression than the promotion of survival. That diffuse overexpression of constitutively active CaMKII strongly promotes RGC survival after axon injury may be clinically valuable for neuroprotection per se. However, the associated strong suppression of the optic nerve axon regeneration demonstrates the need for understanding the intracellular domain- and target-specific CaMKII activities to the development of CaMKII signaling pathway-directed strategies for the treatment of optic neuropathies.

Download

Figure 2. Microglia protect the blood-retina barrier following optic nerve injury (A-C) Retinal flat mounts stained with anti-CD31 and anti-Iba1 from naive and 1 dpc controls and PLX5622-fed mice. Arrowheads mark perivascular, enveloping microglia. Scale bar, 50 mm. (D) Quantification of microglia ablation. y axis, Iba1 + cells per mm 2 . Results are presented as mean ± SEM. Student's t test. **p % 0.01 and ****p % 0.0001. (E) Enveloping microglia in naive (n = 5) and 1 dpc (n = 6) retinas. Results are presented as mean ± SEM. Student's t test. ***p % 0.001. (F) Quantification of BRB leakiness. NaFL entry into the retina, normalized to NaFL in blood. y axis, uptake ratio. N = 3-7 per group (2 retinas each). Data were analyzed with one-way ANOVA followed by Tukey's multiple comparisons. **p % 0.01 and ****p % 0.0001. (G) Quantification of RBPMS + RGCs of naive and 14 dpc mice. Results are presented as mean ± SEM. Data were analyzed with one-way ANOVA followed by Tukey's multiple comparisons. ****p % 0.0001, ns, not significant. See also Figure S3.
Figure 6. Disruption of the classical complement cascade does not enhance axon regeneration (A) Schematic of workflow for analysis of vitreous proteome. (B) Volcano plot of differentially abundant proteins in the Itgam À/À versus WT vitreous. y axis is the Àlog(adjusted) p value and x axis the log2 fold change. (C) Pathway analysis identifies complement and coagulation cascades (purple). (D-G) Feature plots of (D and F) C1q and (E and G) C3 expression in ocular immune cells of WT and Itgam À/À mice. (H-J) CTB-traced axons at 14 dpc and i.o. b-glucan of (H) WT (n = 24 nerves), (I) C1q À/À (n = 8), and (J) C3 À/À (n = 8). Scale bar, 200 mm. (K) Quantification of regenerated axons. Results are presented as mean ± SEM. Data were analyzed with two-way ANOVA followed by Tukey's multiple comparisons. None of the comparisons showed a p value % 0.05. See also Figure S7 and Tables S1 and S2.
Neutrophil-inflicted vasculature damage suppresses immune-mediated optic nerve regeneration

March 2024

·

52 Reads

·

4 Citations

Cell Reports


Figure 2. Inflammatory cell-derived oncomodulin (Ocm) promotes optic nerve regeneration. (A,B) intravitreal injection of zymosan leads to infiltration of neutrophils (red in (A), Gr1 immunostaining) expressing Ocm (green in (B)). (C,D) Ocm immunostaining in the inner retina following intravitreal zymosan injection (GCL: ganglion cell layer; IPL: inner plexiform layer; INL: inner nuclear layer). (E,F) slow-release microspheres loaded with Ocm and CPT-cAMP (Ocm+cAMP) injected into the vitreous induce optic nerve regeneration. Asterisks: injury site. (from refs. [18,22]. Panels (E,F) reprinted with permission from ref. [18]. Copyright 2006, Springer Nature).
Figure 8. Identification of ArmC10 as a high-affinity oncomodulin receptor. (A) identification of a candidate receptor. Using a variant form of proximity biotinylation, lightly fixed RGCs in culture were incubated with either an Fc control probe or an Fc-Ocm fusion protein ("bait"), rinsed, and then exposed to an anti-Fc antibody conjugated to the oxidizing enzyme horseradish peroxidase (HRP). Biotinylation was carried out using the biotin donor (biotin tyramide) in the presence of H2O2. Proteins were extracted from lysed cells and biotinylated proteins were adsorbed onto streptavidin beads that were then separated by SDS-PAGE. Following enzymatic digestion of proteins, peptides were analyzed by mass spectrometry. After excluding obvious impurities, ArmC10 emerged as the protein with the highest enrichment in samples probed with Fc-Ocm vs. Fc. (B) gainof-function studies. After identifying a cell line with low endogenous ArmC10 expression, cells were transfected with a control plasmid or a plasmid encoding ArmC10, then incubated with recombinant Ocm and immunostained to visualize levels of ArmC10 and Ocm. Note elevated Ocm binding following transfection with the ArmC10 plasmid. (C) endogenous ArmC10 expression in wholemounted retina immunostained for III tubulin (to visualize RGCs) and ArmC10. (D) loss-of-function studies. Virally mediated deletion of ArmC10 in RGCs (intraocular AAV2-sgArmC10 + AAV2-Cas9) diminishes axon regeneration induced by Zymosan + CPT-cAMP. (E) addition of the SDF1 antagonist AMD3100 eliminates Zymosan-induced regeneration almost completely. * p < 0.05; *** p < 0.001. (Reprinted with permission from ref. [49]. Copyright 2023, American Academy of Arts and Sciences).
Figure 9. Role of Ocm, ArmC10 and SDF1 in axon regeneration in the sciatic nerve and spinal cord. (A) upper row: Sections through normal adult mouse dorsal root ganglion (DRG) show little monocyte infiltration (F4/80 immunostaining, red) or Ocm (green). Large nuclei of principle sensory neurons are visualized in the last panel with DAPI (blue). Lower row: peripheral nerve injury causes an influx of F4/80-positive monocytes that express Ocm into the DRG. (B) sensory neurons (stained with antibody TUJ1 for βIII tubulin, red) express the Ocm receptor ArmC1 (green). (C) neurite outgrowth from DRG sensory neurons in culture. Treatment with forskolin, mannose, and Ocm (F + M + Ocm)
Figure 11. Accumulation of reactive zinc in amacrine cell terminals after optic nerve injury: another impediment to regeneration. (A) increase in autometallographic (AMG) signal presumed to reflect an elevation of reactive zinc (Zn 2+ ) in synaptic terminals of amacrine cells in the inner plexiform layer (arrows) of the retina one day after optic nerve injury. (B) chelation of Zn 2+ with ZX1 promotes axon regeneration beyond the site visualized two weeks after optic nerve injury. Asterisks denote the injury site (Reprinted with permission from ref. [116], copyright 2017, National Academy of Sciences).
Inflammatory Mediators of Axon Regeneration in the Central and Peripheral Nervous Systems

October 2023

·

81 Reads

·

6 Citations

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.


FEAST: A flow cytometry-based toolkit for interrogating microglial engulfment of synaptic and myelin proteins

September 2023

·

185 Reads

·

8 Citations

Although engulfment is a hallmark of microglia function, fully validated platforms that facilitate high-throughput quantification of this process are lacking. Here, we present FEAST (Flow cytometric Engulfment Assay for Specific Target proteins), which enables interrogation of in vivo engulfment of synaptic material by brain resident macrophages at single-cell resolution. We optimize FEAST for two different analyses: quantification of fluorescent material inside live cells and of engulfed endogenous proteins within fixed cells. To overcome false-positive engulfment signals, we introduce an approach suitable for interrogating engulfment in microglia from perfusion-fixed tissue. As a proof-of-concept for the specificity and versatility of FEAST, we examine the engulfment of synaptic proteins after optic nerve crush and of myelin in two mouse models of demyelination (treatment with cuprizone and injections of lysolecithin). We find that microglia, but not brain-border associated macrophages, engulf in these contexts. Our work underscores how FEAST can be utilized to gain critical insight into functional neuro-immune interactions that shape development, homeostasis, and disease.


Fig. 2 RGC transplantation models, methods, and assessment. Each animal and disease/injury model possesses advantages and disadvantages for studying essential aspects of RGC replacement and mimicking different characteristics of optic neuropathies. Donor RGCs can be delivered to the intravitreal (IVT) or subretinal (SR) space, but each route has unique barriers to overcome to achieve structural integration. In addition to integrating within the host retina, donor RGCs must avoid being targeted by the adaptive and innate immune systems. Visualizing donor and host RGCs is essential to translate cell replacement therapies to the clinic, and a combination of techniques is required to properly assess the structural and functional integration of the transplanted cells
Fig. 3 RGC neurocircuitries in healthy, diseased, and transplanted retinas. Bipolar and amacrine cells establish direct contact with RGCs to relay visual information. Different RGC subtypes extend their dendrites into ON and OFF sublamina in the inner plexiform layer and exhibit different electrophysiological responses. Glaucoma causes dendrite retraction and eventual death of RGCs and the activation of astrocytes, microglia, and Müller glia, while photoreceptor, bipolar, amacrine, and horizontal cells are relatively unaffected. RGC transplantation must replace lost RGCs, return the diseased retina to a homeostatic state, and establish neurocircuitry between host and donor cells. While donor RGCs have been shown to survive in the retina, few are currently able to migrate into the ganglion cell layer, with the inner limiting membrane (ILM) serving as a major barrier for intravitreal (IVT) delivery, and even fewer form de novo neurocircuits in the retina
Retinal ganglion cell repopulation for vision restoration in optic neuropathy: a roadmap from the RReSTORe Consortium

September 2023

·

921 Reads

·

29 Citations

Molecular Neurodegeneration

Retinal ganglion cell (RGC) death in glaucoma and other optic neuropathies results in irreversible vision loss due to the mammalian central nervous system’s limited regenerative capacity. RGC repopulation is a promising therapeutic approach to reverse vision loss from optic neuropathies if the newly introduced neurons can reestablish functional retinal and thalamic circuits. In theory, RGCs might be repopulated through the transplantation of stem cell-derived neurons or via the induction of endogenous transdifferentiation. The RGC Repopulation, Stem Cell Transplantation, and Optic Nerve Regeneration (RReSTORe) Consortium was established to address the challenges associated with the therapeutic repair of the visual pathway in optic neuropathy. In 2022, the RReSTORe Consortium initiated ongoing international collaborative discussions to advance the RGC repopulation field and has identified five critical areas of focus: (1) RGC development and differentiation, (2) Transplantation methods and models, (3) RGC survival, maturation, and host interactions, (4) Inner retinal wiring, and (5) Eye-to-brain connectivity. Here, we discuss the most pertinent questions and challenges that exist on the path to clinical translation and suggest experimental directions to propel this work going forward. Using these five subtopic discussion groups (SDGs) as a framework, we suggest multidisciplinary approaches to restore the diseased visual pathway by leveraging groundbreaking insights from developmental neuroscience, stem cell biology, molecular biology, optical imaging, animal models of optic neuropathy, immunology & immunotolerance, neuropathology & neuroprotection, materials science & biomedical engineering, and regenerative neuroscience. While significant hurdles remain, the RReSTORe Consortium’s efforts provide a comprehensive roadmap for advancing the RGC repopulation field and hold potential for transformative progress in restoring vision in patients suffering from optic neuropathies.


The oncomodulin receptor ArmC10 enables axon regeneration in mice after nerve injury and neurite outgrowth in human iPSC-derived sensory neurons

August 2023

·

108 Reads

·

9 Citations

Science Translational Medicine

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.


Figure 6.
Full-length optic nerve regeneration in the absence of genetic manipulations

February 2023

·

134 Reads

·

14 Citations

JCI Insight

The inability of mature retinal ganglion cells (RGCs) to regenerate axons after optic nerve injury can be partially reversed by manipulating cell-autonomous and/or -non-autonomous factors. Although manipulations of cell-non-autonomous factors could have higher translational potential than genetic manipulations of RGCs, they have generally produced lower levels of optic nerve regeneration. Here we report that preconditioning resulting from mild lens injury (conditioning LI, cLI) prior to optic nerve damage induces far greater regeneration than LI after nerve injury or the pro-inflammatory agent zymosan given either before or after nerve damage. Unlike zymosan-induced regeneration, cLI is unaltered by depleting mature neutrophils or T cells or blocking receptors for known inflammation-derived growth factors (Oncomodulin, SDF1, CCL5), and is only partly diminished by suppressing CCR2+ monocyte recruitment. Repeated episodes of LI lead to full-length optic nerve regeneration, and pharmacological removal of local resident macrophages with the colony stimulating factor 1 receptor (CSF1R) inhibitor PLX5622 enables some axons to re-innervate the brain in just 6 weeks, comparable to the results obtained with the most effective genetic manipulations of RGCs. Thus, cell-non-autonomous interventions can induce high levels of optic nerve regeneration, paving the way to uncover potent, translatable therapeutic targets for CNS repair.


Citations (82)


... CREB is a transcription factor that mediates the expression of genes involved in neuronal development, synaptic plasticity, and neuroprotection 11,34 . In the retina, CaMKII-mediated CREB signaling has been implicated in the regulation of retinal ganglion cell (RGC) survival 35,36 . Our findings demonstrated that high glucose conditions lead to aberrant activation of the CaMK2A/CREB pathway, indicating that excessive activation of this pathway can be detrimental in retinal cells. ...

Reference:

CaMK2A/CREB pathway activation is associated with enhanced mitophagy and neuronal apoptosis in diabetic retinopathy
Ca 2+ /Calmodulin-Dependent Protein Kinase II Enhances Retinal Ganglion Cell Survival But Suppresses Axon Regeneration after Optic Nerve Injury

eNeuro

... Although the role of the BRB has been extensively studied in systemic vascular diseases, such as diabetic retinopathy, its involvement in diseases like glaucoma and optic nerve injury has received comparatively less attention (Jo et al. 2019;Chen et al. 2019). Emerging evidence suggests that BRB dysfunction in these conditions leads to increased vascular permeability, immune cell infiltration, and inflammatory responses, all of which exacerbate RGC degeneration and hinder optic nerve regeneration (McMenamin et al. 2019;Tomkins-Netzer et al. 2024;Passino et al. 2024). The breakdown of the BRB not only disrupts retinal homeostasis but also creates a hostile immune microenvironment for RGCs (Xie et al. 2021;Habibi-Kavashkohie et al. 2023). ...

Neutrophil-inflicted vasculature damage suppresses immune-mediated optic nerve regeneration

Cell Reports

... For example, conditioning lumbosacral sensory neurons by sciatic nerve crush injury accelerates axon regeneration after subsequent PNS injury and brings a degree of axon regrowth after dorsal column lesion in the spinal cord (39). This phenomenon is attributable to the entry of inflammatory cells into the injured PNS, which elevates levels of cell-derived growth factors including oncomodulin and SDF-1 (40). The PTEN®mTOR pathway has been manipulated to increase CNS and PNS axon regeneration either by attenuating PTEN activity or downstream interventions that more directly increase mTOR was not certified by peer review) is the author/funder. ...

Inflammatory Mediators of Axon Regeneration in the Central and Peripheral Nervous Systems

... In this study, we developed an iPSC-derived microglia-neuron coculture and flow cytometry method for measuring microglia uptake of synaptic material, which enabled us to conduct a CRISPR-based screen to systematically assess the effect of ASD-risk genes on microglial uptake of synaptic material. Recently published work presented a similar flow cytometry method for measuring microglial uptake of Alexa Fluor-labeled retinal ganglion cell (RGC) material in the lateral geniculate nucleus (LGN) after optic nerve crush 45 . Synapse remodeling of RGC inputs in the LGN is well defined such that internalization of Alexa Fluor-labeled material by microglia in the LGN can be attributed to synapse remodeling 11 . ...

FEAST: A flow cytometry-based toolkit for interrogating microglial engulfment of synaptic and myelin proteins

... The optic nerve is highly vulnerable to damage and is compromised in neurodegenerative diseases like glaucoma, as a result of ocular trauma or in optic neuropathies. While substantial progress has been made in identifying the molecular and cellular events that lead to RGC death and in identifying potential neuroprotective strategies that could maintain the health of RGCs after axonal damage, no FDAapproved therapies have been identified that promote RGC survival after optic nerve injury [1]. Likewise, while many molecules that enhance RGC axon regeneration post-injury have been identified in a variety of experimental systems, none have yet been shown to be clinically effective to regenerate axons in the human eye [2][3][4][5]. ...

Retinal ganglion cell repopulation for vision restoration in optic neuropathy: a roadmap from the RReSTORe Consortium

Molecular Neurodegeneration

... The second question is what regenerative and neuroprotective factors are expressed in those infiltrating immune cells. There is increasing evidence that infiltrating immune cells express regeneration-enhancing cytokines, trophic factors, and other potential regenerative factors such as CNTF 42 , oncomodulin 70,71 , IL-6 72 and SDF-1 (Cxcl12) 73 , which stimulate axonal regenerative signals in RGCs. However, our RNA-seq data showed that expression of these factors is not affected by fluvastatin and MBV. ...

The oncomodulin receptor ArmC10 enables axon regeneration in mice after nerve injury and neurite outgrowth in human iPSC-derived sensory neurons
  • Citing Article
  • August 2023

Science Translational Medicine

... Lens injury has neuroprotective effects on RGCs, promotes axon growth, and partially alleviates inhibitory conditions. Preconditioning with lens injury 2 weeks before optic nerve crush (ONC) increases optic nerve regeneration threefold and enhances RGC survival (58). However, in CNTF or LIF knockout mice, the regenerative effects of lens injury are significantly reduced (55). ...

Full-length optic nerve regeneration in the absence of genetic manipulations

JCI Insight

... The expression Glycoprotein 130 improves repressor element-1 silencing transcription factor-related axon regenerative capacity in peripheral nerves with aging of REST is decreased in neurodegenerative diseases such as Parkinson's disease and Alzheimer's disease (10). On the other hand, REST inhibits axon regeneration (11). REST has multiple roles, including neuroprotection and neurotoxicity, and a previous study identified it as a critical regulator in neural survival (12). ...

Elk-1 regulates retinal ganglion cell axon regeneration after injury

... By comparison, Injured Day 7 versus Uninjured Day 1 GO analysis showed an enrichment of cytoskeletal changes, potentially in response to axon regrowth, which occurs between 2 and 40 dpi in zebrafish [8,9]. Late response GO enrichment also included endopeptidase activity, DNA methylation, immune responses, autophagy, regulation of the actin cytoskeleton and metabolic changes, each of which has been implicated in RGC regeneration and resiliency responses [33][34][35][36][37][38][39][40][41][42]. Looking specifically at genes within the immune, autophagy and regulation of the actin cytoskeleton pathways revealed differential gene expression, with temporal specificity, during the injury response ( Figure 6E-G). ...

Retinal Ganglion Cell Survival and Axon Regeneration after Optic Nerve Injury: Role of Inflammation and Other Factors

... These latter, pro-survival and pro-regenerative roles are consistent with observations in other neural contexts, where ATF3 is part of a core set of pro-survival and regenerative transcriptional regulators (e.g. [67,79,80]; reviewed in [81]). Examination of immune system-related gene expression also revealed a biphasic immune response after optic nerve injury, with early and late components. ...

Transcription factor network analysis identifies REST/NRSF as an intrinsic regulator of CNS regeneration in mice