Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 24(4): 700-713, 2023 04.
Article in English | MEDLINE | ID: mdl-36807640

ABSTRACT

Non-neuronal cells are key to the complex cellular interplay that follows central nervous system insult. To understand this interplay, we generated a single-cell atlas of immune, glial and retinal pigment epithelial cells from adult mouse retina before and at multiple time points after axonal transection. We identified rare subsets in naive retina, including interferon (IFN)-response glia and border-associated macrophages, and delineated injury-induced changes in cell composition, expression programs and interactions. Computational analysis charted a three-phase multicellular inflammatory cascade after injury. In the early phase, retinal macroglia and microglia were reactivated, providing chemotactic signals concurrent with infiltration of CCR2+ monocytes from the circulation. These cells differentiated into macrophages in the intermediate phase, while an IFN-response program, likely driven by microglia-derived type I IFN, was activated across resident glia. The late phase indicated inflammatory resolution. Our findings provide a framework to decipher cellular circuitry, spatial relationships and molecular interactions following tissue injury.


Subject(s)
Macrophages , Retina , Animals , Mice , Retina/injuries , Retina/metabolism , Microglia , Central Nervous System , Monocytes
2.
Nature ; 624(7991): 403-414, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38092914

ABSTRACT

The brain controls nearly all bodily functions via spinal projecting neurons (SPNs) that carry command signals from the brain to the spinal cord. However, a comprehensive molecular characterization of brain-wide SPNs is still lacking. Here we transcriptionally profiled a total of 65,002 SPNs, identified 76 region-specific SPN types, and mapped these types into a companion atlas of the whole mouse brain1. This taxonomy reveals a three-component organization of SPNs: (1) molecularly homogeneous excitatory SPNs from the cortex, red nucleus and cerebellum with somatotopic spinal terminations suitable for point-to-point communication; (2) heterogeneous populations in the reticular formation with broad spinal termination patterns, suitable for relaying commands related to the activities of the entire spinal cord; and (3) modulatory neurons expressing slow-acting neurotransmitters and/or neuropeptides in the hypothalamus, midbrain and reticular formation for 'gain setting' of brain-spinal signals. In addition, this atlas revealed a LIM homeobox transcription factor code that parcellates the reticulospinal neurons into five molecularly distinct and spatially segregated populations. Finally, we found transcriptional signatures of a subset of SPNs with large soma size and correlated these with fast-firing electrophysiological properties. Together, this study establishes a comprehensive taxonomy of brain-wide SPNs and provides insight into the functional organization of SPNs in mediating brain control of bodily functions.


Subject(s)
Brain , Gene Expression Profiling , Neural Pathways , Neurons , Spinal Cord , Animals , Mice , Hypothalamus , Neurons/metabolism , Neuropeptides , Spinal Cord/cytology , Spinal Cord/metabolism , Brain/cytology , Brain/metabolism , Neurotransmitter Agents , Mesencephalon/cytology , Reticular Formation/cytology , Electrophysiology , Cerebellum/cytology , Cerebral Cortex/cytology
3.
Nature ; 587(7835): 613-618, 2020 11.
Article in English | MEDLINE | ID: mdl-33029008

ABSTRACT

Spinal cord injury in mammals is thought to trigger scar formation with little regeneration of axons1-4. Here we show that a crush injury to the spinal cord in neonatal mice leads to scar-free healing that permits the growth of long projecting axons through the lesion. Depletion of microglia in neonatal mice disrupts this healing process and stalls the regrowth of axons, suggesting that microglia are critical for orchestrating the injury response. Using single-cell RNA sequencing and functional analyses, we find that neonatal microglia are transiently activated and have at least two key roles in scar-free healing. First, they transiently secrete fibronectin and its binding proteins to form bridges of extracellular matrix that ligate the severed ends of the spinal cord. Second, neonatal-but not adult-microglia express several extracellular and intracellular peptidase inhibitors, as well as other molecules that are involved in resolving inflammation. We transplanted either neonatal microglia or adult microglia treated with peptidase inhibitors into spinal cord lesions of adult mice, and found that both types of microglia significantly improved healing and axon regrowth. Together, our results reveal the cellular and molecular basis of the nearly complete recovery of neonatal mice after spinal cord injury, and suggest strategies that could be used to facilitate scar-free healing in the adult mammalian nervous system.


Subject(s)
Microglia/physiology , Spinal Cord Injuries/therapy , Spinal Cord Regeneration , Spinal Cord/cytology , Spinal Cord/physiology , Animals , Animals, Newborn , Axons/drug effects , Axons/physiology , Cicatrix , Fibronectins/metabolism , Homeostasis , Mice , Microglia/drug effects , Protease Inhibitors/pharmacology , RNA-Seq , Single-Cell Analysis , Spinal Cord/pathology , Spinal Cord Injuries/pathology , Spinal Cord Regeneration/drug effects , Wound Healing/drug effects
4.
Klin Monbl Augenheilkd ; 237(2): 133-139, 2020 Feb.
Article in German | MEDLINE | ID: mdl-32040976

ABSTRACT

Glaucoma is a neurodegenerative disease that leads to irreversible blindness over time. Its defining feature is the loss of retinal ganglion cells (RGCs) in the eye and their axons in the optic nerve. Increased intraocular pressure (IOP) is a major risk factor for the development of glaucoma, but is neither necessary nor sufficient for the disease and its progression; this motivates research and development of new strategies for the detection and treatment of glaucoma that focus on neuroprotection - protection of RGCs from dying. In addition, for diagnosis and treatment by reducing IOP, new approaches have been developed in recent years. This article reviews current theories of pathophysiological mechanisms underlying glaucoma and recent research - with a focus on neuroprotection and current preclinical and clinical studies to improve the diagnosis and treatment of glaucoma.


Subject(s)
Glaucoma , Neurodegenerative Diseases , Humans , Intraocular Pressure , Neuroprotection , Optic Nerve , Retinal Ganglion Cells
5.
EMBO J ; 34(9): 1231-43, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25766255

ABSTRACT

The remodeling of axonal circuits after injury requires the formation of new synaptic contacts to enable functional recovery. Which molecular signals initiate such axonal and synaptic reorganisation in the adult central nervous system is currently unknown. Here, we identify FGF22 as a key regulator of circuit remodeling in the injured spinal cord. We show that FGF22 is produced by spinal relay neurons, while its main receptors FGFR1 and FGFR2 are expressed by cortical projection neurons. FGF22 deficiency or the targeted deletion of FGFR1 and FGFR2 in the hindlimb motor cortex limits the formation of new synapses between corticospinal collaterals and relay neurons, delays their molecular maturation, and impedes functional recovery in a mouse model of spinal cord injury. These results establish FGF22 as a synaptogenic mediator in the adult nervous system and a crucial regulator of synapse formation and maturation during post-injury remodeling in the spinal cord.


Subject(s)
Fibroblast Growth Factors/metabolism , Spinal Cord Injuries/metabolism , Synapses/metabolism , Animals , Axons/physiology , Fibroblast Growth Factors/genetics , Mice, Inbred C57BL , Mice, Knockout , Nerve Regeneration/physiology , Neuronal Plasticity/physiology , Neurons/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Signal Transduction , Spinal Cord Injuries/physiopathology , Synapses/physiology
6.
J Neuroinflammation ; 15(1): 240, 2018 Aug 27.
Article in English | MEDLINE | ID: mdl-30149799

ABSTRACT

Neutrophils are the innate immune system's first line of defense. Neutrophils play a critical role in protecting the host against infectious pathogens, resolving sterile injuries, and mediating inflammatory responses. The granules of neutrophils and their constituent proteins are central to these functions. Although neutrophils may exert a protective role upon acute inflammatory conditions or insults, continued activity of neutrophils in chronic inflammatory diseases can contribute to tissue damage. Neutrophil granule proteins are involved in a number of chronic inflammatory conditions and diseases. However, the functions of these proteins in neuroinflammation and chronic neuroinflammatory diseases, including Alzheimer's disease (AD), remain to be elucidated. In this review, we discuss recent findings from our lab and others that suggest possible functions for neutrophils and the neutrophil granule proteins, CAP37, neutrophil elastase, and cathepsin G, in neuroinflammation, with an emphasis on AD. These findings reveal that neutrophil granule proteins may exert both neuroprotective and neurotoxic effects. Further research should determine whether neutrophil granule proteins are valid targets for therapeutic interventions in chronic neuroinflammatory diseases.


Subject(s)
Alzheimer Disease/pathology , Eosinophil Granule Proteins/metabolism , Neurogenic Inflammation/pathology , Neutrophils/metabolism , Alzheimer Disease/immunology , Animals , Humans , Neurogenic Inflammation/immunology
7.
Histochem Cell Biol ; 144(4): 293-308, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26170148

ABSTRACT

Inflammation is a well-defined factor in Alzheimer's disease (AD). There is a strong need to identify the molecules contributing to neuroinflammation so that therapies can be designed to prevent immune-mediated neurotoxicity. The cationic antimicrobial protein of 37 kDa (CAP37) is an inflammatory mediator constitutively expressed in neutrophils (PMNs). In addition to antibiotic activity, CAP37 exerts immunomodulatory effects on microglia. We hypothesize that CAP37 mediates the neuroinflammation associated with AD. However, PMNs are not customarily associated with the pathology of AD. This study was therefore designed to identify non-neutrophilic source(s) of CAP37 in brains of AD patients. Brain tissues from patients and age-matched controls were analyzed for CAP37 expression using immunohistochemistry (IHC). To determine factors that induce CAP37 in AD, HCN-1A primary human neurons were treated with tumor necrosis factor-alpha (TNF-α) or amyloid ß1-40 (Aß) and analyzed by IHC. Western blotting and quantitative reverse transcription polymerase chain reaction (qRT-PCR) were used to confirm CAP37 expression in neurons and brain tissues. IHC revealed CAP37 in cortical neurons in temporal and parietal lobes as well as CA3 and CA4 hippocampal neurons in patients with AD. CAP37 was found in more neurons in AD patients compared with age-matched controls. qRT-PCR and Western blotting showed an increase in CAP37 transcript and protein in the AD temporal lobe, a brain region that is highly impacted in AD. qRT-PCR observations confirmed CAP37 expression in neurons. TNF-α and Aß increased neuronal expression of CAP37. These findings support our hypothesis that neuronal CAP37 may modulate the neuroinflammatory response in AD.


Subject(s)
Alzheimer Disease/metabolism , Antimicrobial Cationic Peptides/metabolism , Blood Proteins/metabolism , Carrier Proteins/metabolism , Inflammation Mediators/metabolism , Pyramidal Cells/metabolism , Adult , Aged , Aged, 80 and over , Alzheimer Disease/diagnosis , Alzheimer Disease/genetics , Amyloid beta-Peptides/pharmacology , Antimicrobial Cationic Peptides/genetics , Blood Proteins/genetics , Carrier Proteins/genetics , Case-Control Studies , Cells, Cultured , Humans , Male , Parietal Lobe/metabolism , Parietal Lobe/pathology , Peptide Fragments/pharmacology , Primary Cell Culture , Pyramidal Cells/drug effects , Pyramidal Cells/pathology , Temporal Lobe/metabolism , Temporal Lobe/pathology , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation , Young Adult
8.
EMBO Rep ; 14(10): 931-7, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23928811

ABSTRACT

If and how neurons remodel their connections after CNS injury critically influences recovery of function. Here, we investigate the role of the growth-initiating transcription factor STAT3 during remodelling of the injured corticospinal tract (CST). Endogenous STAT3 expression in lesioned cortical projection neurons is transient but can be sustained by viral gene transfer. Sustained activation of STAT3 enhances remodelling of lesioned CST fibres and induces de novo formation of collaterals from unlesioned CST fibres. In a unilateral pyramidotomy paradigm, this recruitment of unlesioned fibres leads to the formation of midline crossing circuits that establish ipsilateral forelimb activation and functional recovery.


Subject(s)
Nerve Regeneration , Pyramidal Tracts/physiology , STAT3 Transcription Factor/metabolism , Spinal Cord Injuries/metabolism , Animals , Gene Deletion , Mice , Mice, Inbred C57BL , Neurons/metabolism , Pyramidal Tracts/metabolism , STAT3 Transcription Factor/genetics , Spinal Cord Injuries/pathology
9.
Methods Mol Biol ; 2636: 1-18, 2023.
Article in English | MEDLINE | ID: mdl-36881292

ABSTRACT

A prevalent feature among neurodegenerative conditions, including axonal injury, is that certain neuronal types are disproportionately affected, while others are more resilient. Identifying molecular features that separate resilient from susceptible populations could reveal potential targets for neuroprotection and axon regeneration. A powerful approach to resolve molecular differences across cell types is single-cell RNA-sequencing (scRNA-seq). scRNA-seq is a robustly scalable approach that enables the parallel sampling of gene expression across many individual cells. Here we present a systematic framework to apply scRNA-seq to track neuronal survival and gene expression changes following axonal injury. Our methods utilize the mouse retina because it is an experimentally accessible central nervous system tissue and its cell types have been comprehensively characterized by scRNA-seq. This chapter will focus on preparing retinal ganglion cells (RGCs) for scRNA-seq and pre-processing of sequencing results.


Subject(s)
Axons , Neuroprotection , Animals , Mice , Neuroprotection/genetics , Nerve Regeneration/genetics , Retinal Ganglion Cells , Sequence Analysis, RNA
10.
EMBO Mol Med ; 15(2): e16111, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36601738

ABSTRACT

Functional recovery following incomplete spinal cord injury (SCI) depends on the rewiring of motor circuits during which supraspinal connections form new contacts onto spinal relay neurons. We have recently identified a critical role of the presynaptic organizer FGF22 for the formation of new synapses in the remodeling spinal cord. Here, we now explore whether and how targeted overexpression of FGF22 can be used to mitigate the severe functional consequences of SCI. By targeting FGF22 expression to either long propriospinal neurons, excitatory interneurons, or a broader population of interneurons, we establish that FGF22 can enhance neuronal rewiring both in a circuit-specific and comprehensive way. We can further demonstrate that the latter approach can restore functional recovery when applied either on the day of the lesion or within 24 h. Our study thus establishes viral gene transfer of FGF22 as a new synaptogenic treatment for SCI and defines a critical therapeutic window for its application.


Subject(s)
Spinal Cord Injuries , Humans , Interneurons/metabolism , Interneurons/pathology , Neurons/metabolism , Spinal Cord/pathology , Spinal Cord Injuries/therapy , Synapses/metabolism
11.
J Biol Chem ; 286(37): 32491-501, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21785167

ABSTRACT

α-Phenyl-N-tert-butylnitrone (PBN), a free radical spin trap, has been shown previously to protect retinas against light-induced neurodegeneration, but the mechanism of protection is not known. Here we report that PBN-mediated retinal protection probably occurs by slowing down the rate of rhodopsin regeneration by inhibiting RPE65 activity. PBN (50 mg/kg) protected albino Sprague-Dawley rat retinas when injected 0.5-12 h before exposure to damaging light at 2,700 lux intensity for 6 h but had no effect when administered after the exposure. PBN injection significantly inhibited in vivo recovery of rod photoresponses and the rate of recovery of functional rhodopsin photopigment. Assays for visual cycle enzyme activities indicated that PBN inhibited one of the key enzymes of the visual cycle, RPE65, with an IC(50) = 0.1 mm. The inhibition type for RPE65 was found to be uncompetitive with K(i) = 53 µm. PBN had no effect on the activity of other visual cycle enzymes, lecithin retinol acyltransferase and retinol dehydrogenases. Interestingly, a more soluble form of PBN, N-tert-butyl-α-(2-sulfophenyl) nitrone, which has similar free radical trapping activity, did not protect the retina or inhibit RPE65 activity, providing some insight into the mechanism of PBN specificity and action. Slowing down the visual cycle is considered a treatment strategy for retinal diseases, such as Stargardt disease and dry age-related macular degeneration, in which toxic byproducts of the visual cycle accumulate in retinal cells. Thus, PBN inhibition of RPE65 catalytic action may provide therapeutic benefit for such retinal diseases.


Subject(s)
Carrier Proteins/metabolism , Cyclic N-Oxides/pharmacology , Eye Proteins/metabolism , Light/adverse effects , Neuroprotective Agents/pharmacology , Retinal Degeneration , Retinal Rod Photoreceptor Cells/enzymology , Rhodopsin/metabolism , cis-trans-Isomerases/metabolism , Acyltransferases/metabolism , Alcohol Oxidoreductases/metabolism , Animals , Rats , Rats, Sprague-Dawley , Retinal Degeneration/enzymology , Retinal Degeneration/prevention & control
12.
Article in English | MEDLINE | ID: mdl-34518340

ABSTRACT

Axons are a unique cellular structure that allows for the communication between neurons. Axon damage compromises neuronal communications and often leads to functional deficits. Thus, developing strategies that promote effective axon regeneration for functional restoration is highly desirable. One fruitful approach is to dissect the regenerative mechanisms used by some types of neurons in both mammalian and nonmammalian systems that exhibit spontaneous regenerative capacity. Additionally, numerous efforts have been devoted to deciphering the barriers that prevent successful axon regeneration in the most regeneration-refractory system-the adult mammalian central nervous system. As a result, several regeneration-promoting strategies have been developed, but significant limitations remain. This review is aimed to summarize historic progression and current understanding of this exciting yet incomplete endeavor.


Subject(s)
Axons , Nerve Regeneration , Animals , Axons/physiology , Central Nervous System , Mammals , Nerve Regeneration/physiology , Neurons/physiology
13.
Neuron ; 110(16): 2625-2645.e7, 2022 08 17.
Article in English | MEDLINE | ID: mdl-35767994

ABSTRACT

Injured neurons in the adult mammalian central nervous system often die and seldom regenerate axons. To uncover transcriptional pathways that could ameliorate these disappointing responses, we analyzed three interventions that increase survival and regeneration of mouse retinal ganglion cells (RGCs) following optic nerve crush (ONC) injury, albeit not to a clinically useful extent. We assessed gene expression in each of 46 RGC types by single-cell transcriptomics following ONC and treatment. We also compared RGCs that regenerated with those that survived but did not regenerate. Each intervention enhanced survival of most RGC types, but type-independent axon regeneration required manipulation of multiple pathways. Distinct computational methods converged on separate sets of genes selectively expressed by RGCs likely to be dying, surviving, or regenerating. Overexpression of genes associated with the regeneration program enhanced both survival and axon regeneration in vivo, indicating that mechanistic analysis can be used to identify novel therapeutic strategies.


Subject(s)
Optic Nerve Injuries , Retinal Ganglion Cells , Animals , Axons/metabolism , Cell Survival/genetics , Mammals , Mice , Nerve Regeneration/genetics , Optic Nerve Injuries/genetics , Optic Nerve Injuries/metabolism , Retinal Ganglion Cells/physiology
14.
Biomolecules ; 12(10)2022 09 27.
Article in English | MEDLINE | ID: mdl-36291595

ABSTRACT

Alzheimer's disease (AD) is a multifactorial disease with a complex pathogenesis. Developing multitarget drugs could be a powerful strategy to impact the progressive loss of cognitive functions in this disease. The purpose of this study is to select a multitarget lead peptide candidate among a series of peptide variants derived from the neutrophil granule protein cathepsin G. We screened eight peptide candidates using the following criteria: (1) Inhibition and reversion of amyloid beta (Aß) oligomers, quantified using an enzyme-linked immunosorbent assay (ELISA); (2) direct binding of peptide candidates to the human receptor for advanced glycation end-products (RAGE), the Toll-like receptor 4 (TLR4) and the S100 calcium-binding protein A9 (S100A9), quantified by ELISA; (3) protection against Aß oligomer-induced neuronal cell death, using trypan blue to measure cell death in a murine neuronal cell line; (4) inhibition of TLR4 activation by S100A9, using a human TLR4 reporter cell line. We selected a 27-mer lead peptide that fulfilled these four criteria. This lead peptide is a privileged structure that displays inherent multitarget activity. This peptide is expected to significantly impact cognitive decline in mouse models of Alzheimer's disease, by targeting both neuroinflammation and neurodegeneration.


Subject(s)
Alzheimer Disease , Animals , Mice , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Toll-Like Receptor 4/metabolism , Receptor for Advanced Glycation End Products/metabolism , Cathepsin G/metabolism , Trypan Blue , Calcium-Binding Proteins
15.
Cell Rep ; 40(2): 111040, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35830791

ABSTRACT

Classification and characterization of neuronal types are critical for understanding their function and dysfunction. Neuronal classification schemes typically rely on measurements of electrophysiological, morphological, and molecular features, but aligning such datasets has been challenging. Here, we present a unified classification of mouse retinal ganglion cells (RGCs), the sole retinal output neurons. We use visually evoked responses to classify 1,859 mouse RGCs into 42 types. We also obtain morphological or transcriptomic data from subsets and use these measurements to align the functional classification to publicly available morphological and transcriptomic datasets. We create an online database that allows users to browse or download the data and to classify RGCs from their light responses using a machine learning algorithm. This work provides a resource for studies of RGCs, their upstream circuits in the retina, and their projections in the brain, and establishes a framework for future efforts in neuronal classification and open data distribution.


Subject(s)
Retina , Retinal Ganglion Cells , Animals , Gene Expression , Mice , Retina/physiology , Retinal Ganglion Cells/metabolism
16.
Neuron ; 110(16): 2607-2624.e8, 2022 08 17.
Article in English | MEDLINE | ID: mdl-35767995

ABSTRACT

Regulatory programs governing neuronal death and axon regeneration in neurodegenerative diseases remain poorly understood. In adult mice, optic nerve crush (ONC) injury by severing retinal ganglion cell (RGC) axons results in massive RGC death and regenerative failure. We performed an in vivo CRISPR-Cas9-based genome-wide screen of 1,893 transcription factors (TFs) to seek repressors of RGC survival and axon regeneration following ONC. In parallel, we profiled the epigenetic and transcriptional landscapes of injured RGCs by ATAC-seq and RNA-seq to identify injury-responsive TFs and their targets. These analyses converged on four TFs as critical survival regulators, of which ATF3/CHOP preferentially regulate pathways activated by cytokines and innate immunity and ATF4/C/EBPγ regulate pathways engaged by intrinsic neuronal stressors. Manipulation of these TFs protects RGCs in a glaucoma model. Our results reveal core transcription programs that transform an initial axonal insult into a degenerative process and suggest novel strategies for treating neurodegenerative diseases.


Subject(s)
Optic Nerve Injuries , Retinal Ganglion Cells , Animals , Axons/metabolism , Mice , Mice, Inbred C57BL , Nerve Regeneration/physiology , Optic Nerve Injuries/metabolism , Retinal Ganglion Cells/physiology
17.
Curr Alzheimer Res ; 18(5): 414-427, 2021.
Article in English | MEDLINE | ID: mdl-34429047

ABSTRACT

BACKGROUND: A role for neutrophils in the pathogenesis of Alzheimer's disease (AD) is emerging. We previously showed that the neutrophil granule proteins cationic antimicrobial protein of 37 kDa (CAP37), cathepsin G (CG), and neutrophil elastase (NE) directly bind the amyloid-beta peptide Aß1-42, a central player in AD pathogenesis. CAP37, CG, and NE are serine proteases that can cleave Aß1-42 at different sites and with different catalytic activities. OBJECTIVE: In this study, we compared the effects of these three proteins on Aß1-42 fibrillation and neurotoxicity. METHODS: Using mass spectrometry and in vitro aggregation assay, we found that NE and CG efficiently cleave Aß1-42. This cleavage correlates well with the inhibition of Aß1-42 aggregation into fibrils. In contrast, CAP37 did not efficiently cleave Aß1-42, but was still able to inhibit its fibrillation, most likely through a quenching effect. Inhibition of Aß1-42 aggregation by NE and CG neutralized its toxicity measured in cultured neurons. In contrast, inhibition of Aß1-42 aggregation by CAP37 did not inhibit its neurotoxicity. RESULTS: We found that a peptide derived from CAP37 could mimic the quenching and inhibition of Aß1-42 aggregation effects of the full-length protein. Additionally, this peptide was able to inhibit the neurotoxicity of the most toxic Aß1-42 aggregate, an effect that was not found with the full-length CAP37. CONCLUSION: These results shed light on the mechanisms of action of neutrophil granule proteins with regard to inhibition of Aß1-42 aggregation and neurotoxicity and open up a possible strategy for the discovery of new disease-modifying drugs for AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Neutrophils/metabolism , Peptide Fragments/metabolism , Alzheimer Disease/metabolism , Animals , Antimicrobial Cationic Peptides/metabolism , Blood Proteins/metabolism , Cathepsin G/metabolism , Humans , In Vitro Techniques , Leukocyte Elastase/metabolism , Mice
18.
Sci Rep ; 10(1): 10953, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32616790

ABSTRACT

Anatomically incomplete spinal cord injuries can be followed by functional recovery mediated, in part, by the formation of intraspinal detour circuits. Here, we show that adult mice recover tactile and proprioceptive function following a unilateral dorsal column lesion. We therefore investigated the basis of this recovery and focused on the plasticity of the dorsal column-medial lemniscus pathway. We show that ascending dorsal root ganglion (DRG) axons branch in the spinal grey matter and substantially increase the number of these collaterals following injury. These sensory fibers exhibit synapsin-positive varicosities, indicating their integration into spinal networks. Using a monosynaptic circuit tracing with rabies viruses injected into the cuneate nucleus, we show the presence of spinal cord neurons that provide a detour pathway to the original target area of DRG axons. Notably the number of contacts between DRG collaterals and those spinal neurons increases by more than 300% after injury. We then characterized these interneurons and showed that the lesion triggers a remodeling of the connectivity pattern. Finally, using re-lesion experiments after initial remodeling of connections, we show that these detour circuits are responsible for the recovery of tactile and proprioceptive function. Taken together our study reveals that detour circuits represent a common blueprint for axonal rewiring after injury.


Subject(s)
Ganglia, Spinal/physiology , Nerve Regeneration , Neural Pathways , Neurons/physiology , Recovery of Function , Sensory Receptor Cells/physiology , Spinal Cord Injuries/prevention & control , Animals , Behavior, Animal , Ganglia, Spinal/cytology , Mice , Mice, Inbred C57BL , Neuronal Plasticity , Neurons/cytology , Spinal Cord Injuries/etiology , Spinal Cord Injuries/pathology
19.
Invest Ophthalmol Vis Sci ; 61(4): 16, 2020 04 09.
Article in English | MEDLINE | ID: mdl-32298435

ABSTRACT

Purpose: Corneal abrasion is a common eye injury, and its resolution can be seriously complicated by bacterial infection. We showed that topical application of the cationic antimicrobial protein of 37 kDa (CAP37) promotes corneal re-epithelialization in mice, and peptides derived from CAP37 can recapitulate the antibacterial and wound-healing effects of the full-length protein. The current study was designed to identify the molecular mechanisms mediating the wound-healing effect of CAP37 and derived bioactive peptides. Methods: We used a TriCEPS-based, ligand-receptor glycocapture method to identify the binding partners of CAP37 on live human corneal epithelial cells using the hTCEpi cell line. We used an ELISA method to confirm binding with identified partners and test the binding with CAP37-derived peptides. We used a reporter cell line to measure activation of the identified membrane receptor by CAP37 and derived peptides. Results: We pulled down S100 calcium-binding protein A9 (S100A9) as a binding partner of CAP37 and found that CAP37 and four derived peptides encompassing two regions of CAP37 bind S100A9 with high affinities. We found that CAP37 and the S100A9-binding peptides could also directly interact with the Toll-like receptor 4 (TLR4), a known receptor for S100A9. CAP37 and one peptide partially activated TLR4. The other three peptides did not activate TLR4. Finally, we found that CAP37 and all four peptides could inhibit the activation of TLR4 by S100A9. Conclusions: This study identifies a mechanism of action for CAP37 and derived antimicrobial peptides that may restrain inflammatory responses to corneal injury and favor corneal re-epithelialization.


Subject(s)
Antimicrobial Cationic Peptides/therapeutic use , Blood Proteins/therapeutic use , Calgranulin B/pharmacology , Corneal Injuries/drug therapy , Epithelium, Corneal/drug effects , Toll-Like Receptor 4/metabolism , Wound Healing/drug effects , Administration, Ophthalmic , Animals , Calgranulin B/metabolism , Cell Line , Chromatography, Liquid , Corneal Injuries/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Epithelium, Corneal/metabolism , Female , Humans , Mice , Mice, Inbred C57BL , Ophthalmic Solutions , Tandem Mass Spectrometry
20.
Am J Ther ; 16(6): 517-33, 2009.
Article in English | MEDLINE | ID: mdl-19487926

ABSTRACT

The antioxidant activity of L-carnosine (beta-alanyl-L-histidine, bioactivated in ocular tissues) versus N-acetylcarnosine (N-acetyl-beta-alanyl-L-histidine, ocular-targeted small dipeptide molecules) was studied in aqueous solution and in a lipid environment, employing liposomes as a model of lipid membranes. Reactive oxygen species (ROS) were generated by an iron/ascorbate promoter system for induction of lipid peroxidation (LPO). L-carnosine, which is stabilized from enzymatic hydrolysis, operates as a universal aldehyde and ROS scavenger in both aqueous and lipid environments and is effective at preventing ROS-induced damage to biomolecules. Second-generation carnosine analogs bearing the histidyl-hydrazide moiety were synthesized and tested versus L-carnosine for their ability to reverse the glycation process, also known as the Maillard reaction, and reverse the stable intermolecular cross-links, monitored in the glucose-ethylamine Schiff base model, ultimately resulting in the formation of the advanced glycation end products (AGEs) from nonenzymatic glycation, accumulating in numerous body tissues and fluids. The obtained data demonstrate the transglycation properties of the ophthalmically stabilized L-carnosine and L-carnosine histidyl-hydrazide derivatives tested and can be used to decrease or predict the occurrence of long-term complications of AGE formation and improve therapeutically the quality of vision and length of life for diabetes mellitus patients and survivors with early aging. Scientists at Innovative Vision Products, Inc. (IVP), developed lubricant eyedrops designed as a sustained-release 1% N-acetylcarnosine prodrug of L-carnosine. The eyedrops contain a mucoadhesive cellulose-based compound combined with corneal absorption promoters and glycerine in a drug-delivery system. Anti-aging therapeutics with the ophthalmic drug eyedrop formula including N-acetylcarnosine showed efficacy in the nonsurgical treatment of age-related cataracts for enrolled participants in the prospective, randomized, double-masked, placebo-controlled crossover clinical trial after controlling for age, gender, and daily activities. In a cohort in excess of 50,500 various patients seeking cutting-edge medical care, the N-acetylcarnosine topical eyedrops target therapy was demonstrated to have significant efficacy, safety, and good tolerability for the prevention and treatment of visual impairment in this older population with relatively stable patterns of causes for blindness and visual impairment. Overall, accumulated study data demonstrate that the IVP-designed new vision-saving drugs, including N-acetylcarnosine eyedrops, promote health vision and prevent vision disability from senile cataracts, primary open-angle glaucoma, age-related macular degeneration, diabetic retinopathy, and aging. N-acetylcarnosine eyedrop therapy is the crown jewel of the anti-aging medical movement and revolutionizes early detection, treatment, and rejuvenation of aging-related eye-disabling disorders. N-acetylcarnosine, as an innovative medical science tool and component of the home medicine and alternative medicine approaches, has the potential to alleviate visual impairment and its associated social, economic, and political woes for an aging population.


Subject(s)
Antioxidants/therapeutic use , Carnosine/analogs & derivatives , Cataract/drug therapy , Prodrugs/therapeutic use , Administration, Topical , Aged , Aged, 80 and over , Antioxidants/administration & dosage , Carnosine/administration & dosage , Carnosine/therapeutic use , Cross-Over Studies , Delayed-Action Preparations , Double-Blind Method , Female , Glycation End Products, Advanced/antagonists & inhibitors , Humans , Lipid Peroxidation/drug effects , Liposomes , Male , Middle Aged , Ophthalmic Solutions , Oxidative Stress/drug effects , Prodrugs/administration & dosage , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL