Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.189
Filter
1.
Handb Clin Neurol ; 201: 1-17, 2024.
Article in English | MEDLINE | ID: mdl-38697733

ABSTRACT

Peripheral nerves are functional networks in the body. Disruption of these networks induces varied functional consequences depending on the types of nerves and organs affected. Despite the advances in microsurgical repair and understanding of nerve regeneration biology, restoring full functions after severe traumatic nerve injuries is still far from achieved. While a blunted growth response from axons and errors in axon guidance due to physical barriers may surface as the major hurdles in repairing nerves, critical additional cellular and molecular aspects challenge the orderly healing of injured nerves. Understanding the systematic reprogramming of injured nerves at the cellular and molecular levels, referred to here as "hallmarks of nerve injury regeneration," will offer better ideas. This chapter discusses the hallmarks of nerve injury and regeneration and critical points of failures in the natural healing process. Potential pharmacological and nonpharmacological intervention points for repairing nerves are also discussed.


Subject(s)
Nerve Regeneration , Peripheral Nerve Injuries , Humans , Nerve Regeneration/physiology , Peripheral Nerve Injuries/therapy , Peripheral Nerve Injuries/physiopathology , Animals , Peripheral Nerves , Axons/physiology , Axons/pathology
2.
J Cell Biol ; 223(8)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-38713825

ABSTRACT

Whether, to what extent, and how the axons in the central nervous system (CNS) can withstand sudden mechanical impacts remain unclear. By using a microfluidic device to apply controlled transverse mechanical stress to axons, we determined the stress levels that most axons can withstand and explored their instant responses at nanoscale resolution. We found mild stress triggers a highly reversible, rapid axon beading response, driven by actomyosin-II-dependent dynamic diameter modulations. This mechanism contributes to hindering the long-range spread of stress-induced Ca2+ elevations into non-stressed neuronal regions. Through pharmacological and molecular manipulations in vitro, we found that actomyosin-II inactivation diminishes the reversible beading process, fostering progressive Ca2+ spreading and thereby increasing acute axonal degeneration in stressed axons. Conversely, upregulating actomyosin-II activity prevents the progression of initial injury, protecting stressed axons from acute degeneration both in vitro and in vivo. Our study unveils the periodic actomyosin-II in axon shafts cortex as a novel protective mechanism, shielding neurons from detrimental effects caused by mechanical stress.


Subject(s)
Actomyosin , Axons , Stress, Mechanical , Animals , Mice , Actomyosin/metabolism , Axons/metabolism , Axons/pathology , Calcium/metabolism , Cells, Cultured , Nerve Degeneration/pathology , Rats
3.
Acta Neuropathol Commun ; 12(1): 82, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38812004

ABSTRACT

Neurons pose a particular challenge to degradative processes like autophagy due to their long and thin processes. Autophagic vesicles (AVs) are formed at the tip of the axon and transported back to the soma. This transport is essential since the final degradation of the vesicular content occurs only close to or in the soma. Here, we established an in vivo live-imaging model in the rat optic nerve using viral vector mediated LC3-labeling and two-photon-microscopy to analyze axonal transport of AVs. Under basal conditions in vivo, 50% of the AVs are moving with a majority of 85% being transported in the retrograde direction. Transport velocity is higher in the retrograde than in the anterograde direction. A crush lesion of the optic nerve results in a rapid breakdown of retrograde axonal transport while the anterograde transport stays intact over several hours. Close to the lesion site, the formation of AVs is upregulated within the first 6 h after crush, but the clearance of AVs and the levels of lysosomal markers in the adjacent axon are reduced. Expression of p150Glued, an adaptor protein of dynein, is significantly reduced after crush lesion. In vitro, fusion and colocalization of the lysosomal marker cathepsin D with AVs are reduced after axotomy. Taken together, we present here the first in vivo analysis of axonal AV transport in the mammalian CNS using live-imaging. We find that axotomy leads to severe defects of retrograde motility and a decreased clearance of AVs via the lysosomal system.


Subject(s)
Autophagy , Axonal Transport , Optic Nerve , Animals , Axonal Transport/physiology , Optic Nerve/pathology , Optic Nerve/metabolism , Rats , Autophagy/physiology , Optic Nerve Injuries/metabolism , Optic Nerve Injuries/pathology , Male , Axons/pathology , Axons/metabolism , Nerve Degeneration/pathology , Nerve Degeneration/metabolism , Rats, Sprague-Dawley , Female
4.
Cell Death Dis ; 15(5): 343, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38760361

ABSTRACT

The corticospinal tract (CST) is the principal neural pathway responsible for conducting voluntary movement in the vertebrate nervous system. Netrin-1 is a well-known guidance molecule for midline crossing of commissural axons during embryonic development. Families with inherited Netrin-1 mutations display congenital mirror movements (CMM), which are associated with malformations of pyramidal decussation in most cases. Here, we investigated the role of Netrin-1 in CST formation by generating conditional knockout (CKO) mice using a Gfap-driven Cre line. A large proportion of CST axons spread laterally in the ventral medulla oblongata, failed to decussate and descended in the ipsilateral spinal white matter of Ntn1Gfap CKO mice. Netrin-1 mRNA was expressed in the ventral ventricular zone (VZ) and midline, while Netrin-1 protein was transported by radial glial cells to the ventral medulla, through which CST axons pass. The level of transported Netrin-1 protein was significantly reduced in Ntn1Gfap CKO mice. In addition, Ntn1Gfap CKO mice displayed increased symmetric movements. Our findings indicate that VZ-derived Netrin-1 deletion leads to an abnormal trajectory of the CST in the spinal cord due to the failure of CST midline crossing and provides novel evidence supporting the idea that the Netrin-1 signalling pathway is involved in the pathogenesis of CMM.


Subject(s)
Mice, Knockout , Netrin-1 , Pyramidal Tracts , Animals , Netrin-1/metabolism , Netrin-1/genetics , Mice , Pyramidal Tracts/metabolism , Pyramidal Tracts/pathology , Axons/metabolism , Axons/pathology
5.
Acta Neuropathol ; 147(1): 79, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38705966

ABSTRACT

Although human females appear be at a higher risk of concussion and suffer worse outcomes than males, underlying mechanisms remain unclear. With increasing recognition that damage to white matter axons is a key pathologic substrate of concussion, we used a clinically relevant swine model of concussion to explore potential sex differences in the extent of axonal pathologies. At 24 h post-injury, female swine displayed a greater number of swollen axonal profiles and more widespread loss of axonal sodium channels than males. Axon degeneration for both sexes appeared to be related to individual axon architecture, reflected by a selective loss of small caliber axons after concussion. However, female brains had a higher percentage of small caliber axons, leading to more extensive axon loss after injury compared to males. Accordingly, sexual dimorphism in axonal size is associated with more extensive axonal pathology in females after concussion, which may contribute to worse outcomes.


Subject(s)
Axons , Brain Concussion , Disease Models, Animal , Sex Characteristics , Animals , Female , Axons/pathology , Brain Concussion/pathology , Male , Swine , Brain/pathology
6.
Brain ; 147(6): 2069-2084, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38763511

ABSTRACT

The peroxisomal disease adrenoleukodystrophy (X-ALD) is caused by loss of the transporter of very-long-chain fatty acids (VLCFAs), ABCD1. An excess of VLCFAs disrupts essential homeostatic functions crucial for axonal maintenance, including redox metabolism, glycolysis and mitochondrial respiration. As mitochondrial function and morphology are intertwined, we set out to investigate the role of mitochondrial dynamics in X-ALD models. Using quantitative 3D transmission electron microscopy, we revealed mitochondrial fragmentation in corticospinal axons in Abcd1- mice. In patient fibroblasts, an excess of VLCFAs triggers mitochondrial fragmentation through the redox-dependent phosphorylation of DRP1 (DRP1S616). The blockade of DRP1-driven fission by the peptide P110 effectively preserved mitochondrial morphology. Furthermore, mRNA inhibition of DRP1 not only prevented mitochondrial fragmentation but also protected axonal health in a Caenorhabditis elegans model of X-ALD, underscoring DRP1 as a potential therapeutic target. Elevated levels of circulating cell-free mtDNA in patients' CSF align this leukodystrophy with primary mitochondrial disorders. Our findings underscore the intricate interplay between peroxisomal dysfunction, mitochondrial dynamics and axonal integrity in X-ALD, shedding light on potential avenues for therapeutic intervention.


Subject(s)
ATP Binding Cassette Transporter, Subfamily D, Member 1 , Adrenoleukodystrophy , Dynamins , Mitochondrial Dynamics , Adrenoleukodystrophy/metabolism , Adrenoleukodystrophy/pathology , Adrenoleukodystrophy/genetics , Animals , Mitochondrial Dynamics/physiology , Humans , Mice , Dynamins/metabolism , Dynamins/genetics , ATP Binding Cassette Transporter, Subfamily D, Member 1/genetics , Caenorhabditis elegans , Mitochondria/metabolism , Mitochondria/pathology , Axons/pathology , Axons/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Male , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Disease Models, Animal , Pyramidal Tracts/pathology , Pyramidal Tracts/metabolism , Peptide Fragments , GTP Phosphohydrolases
7.
Int Immunopharmacol ; 134: 112188, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38728880

ABSTRACT

Neuroinflammation is one of the extensive secondary injury processes that aggravate metabolic and cellular dysfunction and tissue loss following spinal cord injury (SCI). Thus, an anti-inflammatory strategy is crucial for modulating structural and functional restoration during the stage of acute and chronic SCI. Recombinant fibroblast growth factor 4 (rFGF4) has eliminated its mitogenic activity and demonstrated a metabolic regulator for alleviating hyperglycemia in type 2 diabetes and liver injury in non-alcoholic steatohepatitis. However, it remains to be explored whether or not rFGF4 has a neuroprotective effect for restoring neurological disorders, such as SCI. Here, we identified that rFGF4 could polarize microglia/macrophages into the restorative M2 subtype, thus exerting an anti-inflammatory effect to promote neurological functional recovery and nerve fiber regeneration after SCI. Importantly, these effects by rFGF4 were related to triggering PI3K/AKT/GSK3ß and attenuating TLR4/NF-κB signaling axes. Conversely, gene silencing of the PI3K/AKT/GSK3ß signaling or pharmacological reactivation of the TLR4/NF-κB axis aggravated inflammatory reaction. Thus, our findings highlight rFGF4 as a potentially therapeutic regulator for repairing SCI, and its outstanding effect is associated with regulating macrophage/microglial polarization.


Subject(s)
Glycogen Synthase Kinase 3 beta , Macrophages , Microglia , NF-kappa B , Nerve Regeneration , Recovery of Function , Spinal Cord Injuries , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/drug therapy , Animals , Microglia/drug effects , Microglia/metabolism , Macrophages/drug effects , Macrophages/immunology , Nerve Regeneration/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , NF-kappa B/metabolism , Recombinant Proteins/therapeutic use , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Toll-Like Receptor 4/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Mice , Male , Axons/metabolism , Axons/drug effects , Axons/pathology , Proto-Oncogene Proteins c-akt/metabolism , Mice, Inbred C57BL , Rats, Sprague-Dawley , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Phenotype , Rats , Humans , Disease Models, Animal , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology
8.
Cell Rep Med ; 5(5): 101554, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38729157

ABSTRACT

The axons of retinal ganglion cells (RGCs) form the optic nerve, transmitting visual information from the eye to the brain. Damage or loss of RGCs and their axons is the leading cause of visual functional defects in traumatic injury and degenerative diseases such as glaucoma. However, there are no effective clinical treatments for nerve damage in these neurodegenerative diseases. Here, we report that LIM homeodomain transcription factor Lhx2 promotes RGC survival and axon regeneration in multiple animal models mimicking glaucoma disease. Furthermore, following N-methyl-D-aspartate (NMDA)-induced excitotoxicity damage of RGCs, Lhx2 mitigates the loss of visual signal transduction. Mechanistic analysis revealed that overexpression of Lhx2 supports axon regeneration by systematically regulating the transcription of regeneration-related genes and inhibiting transcription of Semaphorin 3C (Sema3C). Collectively, our studies identify a critical role of Lhx2 in promoting RGC survival and axon regeneration, providing a promising neural repair strategy for glaucomatous neurodegeneration.


Subject(s)
Axons , Disease Models, Animal , Glaucoma , LIM-Homeodomain Proteins , Nerve Regeneration , Retinal Ganglion Cells , Transcription Factors , Animals , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Glaucoma/genetics , Glaucoma/pathology , Glaucoma/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Axons/metabolism , Axons/pathology , Mice , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Mice, Inbred C57BL , Cell Survival/genetics , Semaphorins/metabolism , Semaphorins/genetics , N-Methylaspartate/metabolism
9.
J Neuroinflammation ; 21(1): 134, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802868

ABSTRACT

BACKGROUND: Since the 1990s, evidence has accumulated that macrophages promote peripheral nerve regeneration and are required for enhancing regeneration in the conditioning lesion (CL) response. After a sciatic nerve injury, macrophages accumulate in the injury site, the nerve distal to that site, and the axotomized dorsal root ganglia (DRGs). In the peripheral nervous system, as in other tissues, the macrophage response is derived from both resident macrophages and recruited monocyte-derived macrophages (MDMs). Unresolved questions are: at which sites do macrophages enhance nerve regeneration, and is a particular population needed. METHODS: Ccr2 knock-out (KO) and Ccr2gfp/gfp knock-in/KO mice were used to prevent MDM recruitment. Using these strains in a sciatic CL paradigm, we examined the necessity of MDMs and residents for CL-enhanced regeneration in vivo and characterized injury-induced nerve inflammation. CL paradigm variants, including the addition of pharmacological macrophage depletion methods, tested the role of various macrophage populations in initiating or sustaining the CL response. In vivo regeneration, measured from bilateral proximal test lesions (TLs) after 2 d, and macrophages were quantified by immunofluorescent staining. RESULTS: Peripheral CL-enhanced regeneration was equivalent between crush and transection CLs and was sustained for 28 days in both Ccr2 KO and WT mice despite MDM depletion. Similarly, the central CL response measured in dorsal roots was unchanged in Ccr2 KO mice. Macrophages at both the TL and CL, but not between them, stained for the pro-regenerative marker, arginase 1. TL macrophages were primarily CCR2-dependent MDMs and nearly absent in Ccr2 KO and Ccr2gfp/gfp KO mice. However, there were only slightly fewer Arg1+ macrophages in CCR2 null CLs than controls due to resident macrophage compensation. Zymosan injection into an intact WT sciatic nerve recruited Arg1+ macrophages but did not enhance regeneration. Finally, clodronate injection into Ccr2gfp KO CLs dramatically reduced CL macrophages. Combined with the Ccr2gfp KO background, depleting MDMs and TL macrophages, and a transection CL, physically removing the distal nerve environment, nearly all macrophages in the nerve were removed, yet CL-enhanced regeneration was not impaired. CONCLUSIONS: Macrophages in the sciatic nerve are neither necessary nor sufficient to produce a CL response.


Subject(s)
Macrophages , Nerve Regeneration , Peripheral Nerve Injuries , Receptors, CCR2 , Wallerian Degeneration , Animals , Macrophages/metabolism , Macrophages/pathology , Mice , Nerve Regeneration/physiology , Wallerian Degeneration/pathology , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Receptors, CCR2/deficiency , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/metabolism , Mice, Inbred C57BL , Mice, Knockout , Sciatic Neuropathy/pathology , Axons/pathology , Mice, Transgenic , Disease Models, Animal , Sciatic Nerve/injuries , Sciatic Nerve/pathology , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism
10.
Amino Acids ; 56(1): 32, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38637413

ABSTRACT

Diabetic neuropathy (DN) is a common neurological complication caused by diabetes mellitus (DM). Axonal degeneration is generally accepted to be the major pathological change in peripheral DN. Taurine has been evidenced to be neuroprotective in various aspects, but its effect on spinal cord axon injury (SCAI) in DN remains barely reported. This study showed that taurine significantly ameliorated axonal damage of spinal cord (SC), based on morphological and functional analyses, in a rat model of DN induced by streptozotocin (STZ). Taurine was also found to induce neurite outgrowth in cultured cerebral cortex neurons with high glucose exposure. Moreover, taurine up-regulated the expression of nerve growth factor (NGF) and neurite outgrowth relative protein GAP-43 in rat DN model and cultured cortical neurons/VSC4.1 cells. Besides, taurine increased the activating phosphorylation signals of TrkA, Akt, and mTOR. Mechanistically, the neuroprotection by taurine was related to the NGF-pAKT-mTOR axis, because either NGF-neutralizing antibody or Akt or mTOR inhibitors was found to attenuate its beneficial effects. Together, our results demonstrated that taurine promotes spinal cord axon repair in a model of SCAI in STZ-induced diabetic rats, mechanistically associating with the NGF-dependent activation of Akt/mTOR pathway.


Subject(s)
Diabetes Mellitus, Experimental , Proto-Oncogene Proteins c-akt , Animals , Rats , Axons/metabolism , Axons/pathology , Diabetes Mellitus, Experimental/metabolism , Nerve Growth Factor/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Taurine/pharmacology , Taurine/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
11.
Clinics (Sao Paulo) ; 79: 100359, 2024.
Article in English | MEDLINE | ID: mdl-38657346

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate the GSH effect on functional and histological recovery after experimental spinal cord injury in rats. METHODS: Forty Wistar rats were subjected to spinal cord injury through the Multicenter Animal Spinal Cord Injury Study (MASCIS) Impactor system. The rats were sorted and divided into four groups, as follows: Group 1 ‒ Laminectomy and spinal cord injury; Group 2 ‒ Laminectomy, spinal cord injury and Saline Solution (SS) 0.9%; Group 3 ‒ Laminectomy, spinal cord injury, and GSH; and Group 4 ‒ lLaminectomy without spinal cord injury. GSH and SS were administered intraperitoneally. Groups 1 and 4 received no intervention. RESULTS: The rats were evaluated for locomotor function recovery at seven different times by the Basso, Beattie, and Bresnahan (BBB) scale on days 2, 7, 14, 21, 28, 35, and 42 after the spinal cord injury. On day 42, the rats were sacrificed to analyze the histological findings of the injured spinal cord. In the group submitted to GSH, our experimental study revealed better functional scores on the BBB scale, horizontal ladder scale, and cranial and caudal axon count. The differences found were statistically significant in BBB scores and axonal count analysis. CONCLUSION: This study demonstrated that using glutathione in experimental spinal trauma can lead to better functional recovery and improved axonal regeneration rate in Wistar rats submitted to experimental spinal cord injury.


Subject(s)
Disease Models, Animal , Glutathione , Rats, Wistar , Recovery of Function , Spinal Cord Injuries , Animals , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/pathology , Time Factors , Laminectomy , Male , Spinal Cord/pathology , Spinal Cord/physiopathology , Random Allocation , Rats , Axons/pathology , Locomotion/physiology , Reproducibility of Results , Motor Activity/physiology , Treatment Outcome
12.
Int J Mol Sci ; 25(8)2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38673871

ABSTRACT

Mild traumatic brain injury (mTBI) affects millions of people in the U.S. Approximately 20-30% of those individuals develop adverse symptoms lasting at least 3 months. In a rat mTBI study, the closed-head impact model of engineered rotational acceleration (CHIMERA) produced significant axonal injury in the optic tract (OT), indicating white-matter damage. Because retinal ganglion cells project to the lateral geniculate nucleus (LGN) in the thalamus through the OT, we hypothesized that synaptic density may be reduced in the LGN of rats following CHIMERA injury. A modified SEQUIN (synaptic evaluation and quantification by imaging nanostructure) method, combined with immunofluorescent double-labeling of pre-synaptic (synapsin) and post-synaptic (PSD-95) markers, was used to quantify synaptic density in the LGN. Microglial activation at the CHIMERA injury site was determined using Iba-1 immunohistochemistry. Additionally, the effects of ketamine, a potential neuroprotective drug, were evaluated in CHIMERA-induced mTBI. A single-session repetitive (ssr-) CHIMERA (3 impacts, 1.5 joule/impact) produced mild effects on microglial activation at the injury site, which was significantly enhanced by post-injury intravenous ketamine (10 mg/kg) infusion. However, ssr-CHIMERA did not alter synaptic density in the LGN, although ketamine produced a trend of reduction in synaptic density at post-injury day 4. Further research is necessary to characterize the effects of ssr-CHIMERA and subanesthetic doses of intravenous ketamine on different brain regions and multiple time points post-injury. The current study demonstrates the utility of the ssr-CHIMERA as a rodent model of mTBI, which researchers can use to identify biological mechanisms of mTBI and to develop improved treatment strategies for individuals suffering from head trauma.


Subject(s)
Ketamine , Microglia , Rats, Sprague-Dawley , Synapses , Animals , Ketamine/administration & dosage , Ketamine/pharmacology , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Rats , Male , Synapses/drug effects , Synapses/metabolism , Synapses/pathology , Head Injuries, Closed/pathology , Axons/drug effects , Axons/metabolism , Axons/pathology , Disease Models, Animal , Geniculate Bodies/pathology , Geniculate Bodies/drug effects , Brain Concussion/pathology , Brain Concussion/metabolism , Disks Large Homolog 4 Protein/metabolism , Synapsins/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/administration & dosage
13.
Brain Nerve ; 76(4): 361-374, 2024 Apr.
Article in Japanese | MEDLINE | ID: mdl-38589281

ABSTRACT

Recent advances in genetic and antibody testing have limited pathological examination of peripheral nerve specimens. However, when examining peripheral neuropathological findings from a modern perspective, there is often an opportunity to comprehend previously unnoticed observations upon re-examining the same specimen. For example, electron microscopy studies have suggested that the components that distinguish between nodal regions and internodes play a pivotal role in the behavior of macrophages that initiate myelin phagocytosis in the demyelinating form of Guillain-Barré syndrome and chronic inflammatory demyelinating polyneuropathy (CIDP). Conversely, some patients previously diagnosed with CIDP were found to possess distinctive mechanisms initiated by autoantibodies against paranodal junction proteins such as neurofascin 155 leading to the emergence of the concept of autoimmune nodopathy. In vasculitis, the roles of neutrophils in antineutrophil cytoplasmic antibody-associated vasculitis, eosinophils in eosinophilic granulomatosis with polyangiitis, and complements in nonsystemic vasculitic neuropathy in tissue damage have also been demonstrated when viewed from a modern perspective. Furthermore, mechanisms attributable to predominant small-fiber loss in hereditary transthyretin amyloidosis have been clarified at an ultrastructural level.


Subject(s)
Churg-Strauss Syndrome , Granulomatosis with Polyangiitis , Nervous System Diseases , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating , Humans , Churg-Strauss Syndrome/pathology , Granulomatosis with Polyangiitis/pathology , Axons/pathology , Autoantibodies
14.
Transl Psychiatry ; 14(1): 194, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649377

ABSTRACT

Recent research has highlighted the role of complement genes in shaping the microstructure of the brain during early development, and in contributing to common allele risk for Schizophrenia. We hypothesised that common risk variants for schizophrenia within complement genes will associate with structural changes in white matter microstructure within tracts innervating the frontal lobe. Results showed that risk alleles within the complement gene set, but also intergenic alleles, significantly predict axonal density in white matter tracts connecting frontal cortex with parietal, temporal and occipital cortices. Specifically, risk alleles within the Major Histocompatibility Complex region in chromosome 6 appeared to drive these associations. No significant associations were found for the orientation dispersion index. These results suggest that changes in axonal packing - but not in axonal coherence - determined by common risk alleles within the MHC genomic region - including variants related to the Complement system - appear as a potential neurobiological mechanism for schizophrenia.


Subject(s)
Alleles , Genetic Predisposition to Disease , Major Histocompatibility Complex , Schizophrenia , White Matter , Humans , Schizophrenia/genetics , Schizophrenia/pathology , White Matter/pathology , White Matter/diagnostic imaging , Female , Male , Adult , Major Histocompatibility Complex/genetics , Young Adult , Frontal Lobe/pathology , Frontal Lobe/diagnostic imaging , Middle Aged , Diffusion Tensor Imaging , Chromosomes, Human, Pair 6/genetics , Axons/pathology , Polymorphism, Single Nucleotide
15.
Orphanet J Rare Dis ; 19(1): 138, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38549180

ABSTRACT

Although the pathology of X-linked adrenoleukodystrophy (ALD) is well described, it represents the end-stage of neurodegeneration. It is still unclear what cell types are initially involved and what their role is in the disease process. Revisiting the seminal post-mortem studies from the 1970s can generate new hypotheses on pathophysiology. This review describes (histo)pathological changes of the brain and spinal cord in ALD. It aims at integrating older works with current insights and at providing an overarching theory on the pathophysiology of ALD. The data point to an important role for axons and glia in the pathology of both the myelopathy and leukodystrophy of ALD. In-depth pathological analyses with new techniques could help further unravel the sequence of events behind the pathology of ALD.


Subject(s)
Adrenoleukodystrophy , Spinal Cord Diseases , Humans , Adrenoleukodystrophy/pathology , Axons/metabolism , Axons/pathology
16.
Adv Biol (Weinh) ; 8(5): e2400020, 2024 May.
Article in English | MEDLINE | ID: mdl-38548657

ABSTRACT

Understanding the intricate processes of neuronal growth, degeneration, and neurotoxicity is paramount for unraveling nervous system function and holds significant promise in improving patient outcomes, especially in the context of chemotherapy-induced peripheral neuropathy (CIPN). These processes are influenced by a broad range of entwined events facilitated by chemical, electrical, and mechanical signals. The progress of each process is inherently linked to phenotypic changes in cells. Currently, the primary means of demonstrating morphological changes rely on measurements of neurite outgrowth and axon length. However, conventional techniques for monitoring these processes often require extensive preparation to enable manual or semi-automated measurements. Here, a label-free and non-invasive approach is employed for monitoring neuronal differentiation and degeneration using quantitative phase imaging (QPI). Operating on unlabeled specimens and offering little to no phototoxicity and photobleaching, QPI delivers quantitative maps of optical path length delays that provide an objective measure of cellular morphology and dynamics. This approach enables the visualization and quantification of axon length and other physical properties of dorsal root ganglion (DRG) neuronal cells, allowing greater understanding of neuronal responses to stimuli simulating CIPN conditions. This research paves new avenues for the development of more effective strategies in the clinical management of neurotoxicity.


Subject(s)
Axons , Cell Differentiation , Ganglia, Spinal , Animals , Ganglia, Spinal/pathology , Ganglia, Spinal/cytology , Axons/pathology , Neurons/pathology , Humans , Mice , Peripheral Nervous System Diseases/pathology , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/physiopathology , Quantitative Phase Imaging
17.
J Neurol ; 271(6): 3512-3526, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38536455

ABSTRACT

BACKGROUND: Definitions of aggressive MS employ clinical and MR imaging criteria to identify highly active, rapidly progressing disease courses. However, the degree of overlap between clinical and radiological parameters and biochemical markers of CNS injury is not fully understood. Aim of this cross-sectional study was to match clinical and MR imaging hallmarks of aggressive MS to serum/CSF markers of neuroaxonal and astroglial injury (neurofilament light chain (sNfL, cNfL), and glial fibrillary acidic protein (sGFAP, cGFAP)). METHODS: We recruited 77 patients with relapsing-remitting MS (RRMS) and 22 patients with clinically isolated syndrome. NfL and GFAP levels in serum and CSF were assessed using a single-molecule-array HD-1-analyzer. A general linear model with each biomarker as a dependent variable was computed. Clinical and imaging criteria of aggressive MS, as recently proposed by the ECTRIMS Consensus Group, were modeled as independent variables. Other demographic, clinical or laboratory parameters, were modeled as covariates. Analyses were repeated in a homogenous subgroup, consisting only of newly diagnosed, treatment-naïve RRMS patients presenting with an acute relapse. RESULTS: After adjusting for covariates and multiplicity of testing, sNfL and cNfL concentrations were strongly associated with the presence of ≥2 gadolinium-enhancing lesions (psNfL = 0.00008; pcNfL = 0.004) as well as the presence of infratentorial lesions on MRI (psNfL = 0.0003; pcNfL < 0.004). No other clinical and imaging criteria of aggressive MS correlated significantly with NfL or GFAP in serum and CSF. In the more homogeneous subgroup, sNfL still was associated with the presence of ≥2 gadolinium-enhancing lesions (psNfL = 0.001), presence of more than 20 T2-lesions (psNfL = 0.049) as well as the presence of infratentorial lesions on MRI (psNfL = 0.034), while cNfL was associated with the presence of ≥2 gadolinium-enhancing lesions (psNfL = 0.011) and presence of more than 20 T2-lesions (psNfL = 0.029). CONCLUSIONS: Among proposed risk factors for an aggressive disease course, MRI findings but not clinical characteristics correlated with sNfL and cNfL as a marker of neuroaxonal injury and should be given appropriate weight considering MS prognosis and therapy. No significant correlation was detected for GFAP alone.


Subject(s)
Biomarkers , Glial Fibrillary Acidic Protein , Magnetic Resonance Imaging , Neurofilament Proteins , Humans , Male , Female , Adult , Glial Fibrillary Acidic Protein/cerebrospinal fluid , Glial Fibrillary Acidic Protein/blood , Neurofilament Proteins/blood , Neurofilament Proteins/cerebrospinal fluid , Biomarkers/cerebrospinal fluid , Biomarkers/blood , Cross-Sectional Studies , Multiple Sclerosis, Relapsing-Remitting/cerebrospinal fluid , Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Multiple Sclerosis, Relapsing-Remitting/blood , Multiple Sclerosis, Relapsing-Remitting/pathology , Middle Aged , Young Adult , Axons/pathology , Neuroglia/pathology , Demyelinating Diseases/cerebrospinal fluid , Demyelinating Diseases/diagnostic imaging , Demyelinating Diseases/blood
18.
J Biol Chem ; 300(4): 107137, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38447793

ABSTRACT

Experimental studies in flies, mice, and humans suggest a significant role of impaired axonal transport in the pathogenesis of Alzheimer's disease (AD). The mechanisms underlying these impairments in axonal transport, however, remain poorly understood. Here we report that the Swedish familial AD mutation causes a standstill of the amyloid precursor protein (APP) in the axons at the expense of its reduced anterograde transport. The standstill reflects the perturbed directionality of the axonal transport of APP, which spends significantly more time traveling in the retrograde direction. This ineffective movement is accompanied by an enhanced association of dynactin-1 with APP, which suggests that reduced anterograde transport of APP is the result of enhanced activation of the retrograde molecular motor dynein by dynactin-1. The impact of the Swedish mutation on axonal transport is not limited to the APP vesicles since it also reverses the directionality of a subset of early endosomes, which become enlarged and aberrantly accumulate in distal locations. In addition, it also reduces the trafficking of lysosomes due to their less effective retrograde movement. Altogether, our experiments suggest a pivotal involvement of retrograde molecular motors and transport in the mechanisms underlying impaired axonal transport in AD and reveal significantly more widespread derangement of axonal transport pathways in the pathogenesis of AD.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Axonal Transport , Animals , Humans , Mice , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Axonal Transport/genetics , Axons/metabolism , Axons/pathology , Dynactin Complex/metabolism , Dynactin Complex/genetics , Dyneins/metabolism , Endosomes/metabolism , Endosomes/genetics , Lysosomes/metabolism , Mutation , Genetic Variation
19.
J Vis Exp ; (204)2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38465945

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is a common immune-based model of multiple sclerosis (MS). This disease can be induced in rodents by active immunization with protein components of the myelin sheath and Complete Freund's adjuvant (CFA) or by the transfer of myelin-specific T effector cells from rodents primed with myelin protein/CFA into naïve rodents. The severity of EAE is typically scored on a 5-point clinical scale that measures the degree of ascending paralysis, but this scale is not optimal for assessing the extent of recovery from EAE. For example, clinical scores remain high in some EAE models (e.g., myelin oligodendrocyte glycoprotein [MOG] peptide-induced model of EAE) despite the resolution of inflammation. Thus, it is important to complement clinical scoring with histological scoring of EAE, which also provides a means to study the underlying mechanisms of cellular injury in the central nervous system (CNS). Here, a simple protocol is presented to prepare and stain spinal cord and brain sections from mice and to score inflammation, demyelination, and axonal injury in the spinal cord. The method for scoring leukocyte infiltration in the spinal cord can also be applied to score brain inflammation in EAE. A protocol for measuring soluble neurofilament light (sNF-L) in the serum of mice using a Small Molecule Assay (SIMOA) assay is also described, which provides feedback on the extent of overall CNS injury in live mice.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Mice , Animals , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Multiple Sclerosis/pathology , Spinal Cord/pathology , Inflammation/pathology , Axons/pathology , Myelin-Oligodendrocyte Glycoprotein , Mice, Inbred C57BL , Peptide Fragments/adverse effects
20.
Int J Biol Macromol ; 263(Pt 2): 130333, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38408580

ABSTRACT

The cystic cavity that develops following spinal cord injury is a major obstacle for repairing spinal cord injury (SCI). The injectable self-healing biomaterials treatment is a promising strategy to enhance tissue repair after traumatic spinal cord injury. Herein, a natural extracellular matrix (ECM) biopolymer hyaluronic acid-based hydrogel was developed based on multiple dynamic covalent bonds. The hydrogels exhibited excellent injectable and self-healing properties, could be effectively injected into the injury site, and filled the lesion cavity to accelerate the tissue repair of traumatic SCI. Moreover, the hydrogels were compatible with cells and various tissues and possessed proper stiffness matched with nervous tissue. Additionally, when implanted into the injured spinal cord site, the hyaluronic acid-based hydrogel promoted axonal regeneration and functional recovery by accelerating remyelination, axon regeneration, and angiogenesis. Overall, the injectable self-healing hyaluronic acid-based hydrogels are ideal biomaterials for treating traumatic SCI.


Subject(s)
Spinal Cord Injuries , Spinal Cord Regeneration , Humans , Hyaluronic Acid/pharmacology , Hyaluronic Acid/chemistry , Axons/pathology , Hydrogels/chemistry , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/pathology , Biocompatible Materials/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...