Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 36
1.
Elife ; 122024 Mar 26.
Article En | MEDLINE | ID: mdl-38529532

Increased levels of lactate, an end-product of glycolysis, have been proposed as a potential surrogate marker for metabolic changes during neuronal excitation. These changes in lactate levels can result in decreased brain pH, which has been implicated in patients with various neuropsychiatric disorders. We previously demonstrated that such alterations are commonly observed in five mouse models of schizophrenia, bipolar disorder, and autism, suggesting a shared endophenotype among these disorders rather than mere artifacts due to medications or agonal state. However, there is still limited research on this phenomenon in animal models, leaving its generality across other disease animal models uncertain. Moreover, the association between changes in brain lactate levels and specific behavioral abnormalities remains unclear. To address these gaps, the International Brain pH Project Consortium investigated brain pH and lactate levels in 109 strains/conditions of 2294 animals with genetic and other experimental manipulations relevant to neuropsychiatric disorders. Systematic analysis revealed that decreased brain pH and increased lactate levels were common features observed in multiple models of depression, epilepsy, Alzheimer's disease, and some additional schizophrenia models. While certain autism models also exhibited decreased pH and increased lactate levels, others showed the opposite pattern, potentially reflecting subpopulations within the autism spectrum. Furthermore, utilizing large-scale behavioral test battery, a multivariate cross-validated prediction analysis demonstrated that poor working memory performance was predominantly associated with increased brain lactate levels. Importantly, this association was confirmed in an independent cohort of animal models. Collectively, these findings suggest that altered brain pH and lactate levels, which could be attributed to dysregulated excitation/inhibition balance, may serve as transdiagnostic endophenotypes of debilitating neuropsychiatric disorders characterized by cognitive impairment, irrespective of their beneficial or detrimental nature.


Cognitive Dysfunction , Endophenotypes , Animals , Mice , Humans , Brain/metabolism , Cognitive Dysfunction/metabolism , Disease Models, Animal , Lactates/metabolism , Hydrogen-Ion Concentration
2.
Neurosci Res ; 2024 Mar 19.
Article En | MEDLINE | ID: mdl-38508957

Sleep apnea is regarded as an important risk factor in the pathogenesis of Alzheimer disease (AD). Chronic intermittent hypoxia treatment (IHT) given during the sleep period of the circadian cycle in experimental animals is a well-established sleep apnea model. Here we report that transient IHT for 4 days on AD model mice causes Aß overproduction 2 months after IHT presumably via upregulation of synaptic BACE1, side-by-side with tau hyperphosphorylation. These results suggest that even transient IHT may be sufficient to cause long-lasting changes in the molecules measured as AD biomarkers in the brain.

3.
PLoS One ; 18(5): e0285897, 2023.
Article En | MEDLINE | ID: mdl-37224113

Peripheral nerves conducting motor and somatosensory signals in vertebrate consist of myelinated and unmyelinated axons. In vitro myelination culture, generated by co-culturing Schwann cells (SCs) and dorsal root ganglion (DRG) neurons, is an indispensable tool for modeling physiological and pathological conditions of the peripheral nervous system (PNS). This technique allows researchers to overexpress or downregulate molecules investigated in neurons or SCs to evaluate the effect of such molecules on myelination. In vitro myelination experiments are usually time-consuming and labor-intensive to perform. Here we report an optimized protocol for in vitro myelination using DRG explant culture. We found that our in vitro myelination using DRG explant (IVMDE) culture not only achieves myelination with higher efficiency than conventional in vitro myelination methods, but also can be used to observe Remak bundle and non-myelinating SCs, which were unrecognizable in conventional methods. Because of these characteristics, IVMDE may be useful in modeling PNS diseases, including Charcot Marie Tooth disease (CMT), in vitro. These results suggest that IVMDE may achieve a condition more similar to peripheral nerve myelination observed during physiological development.


Ganglia, Spinal , Peripheral Nervous System , Schwann Cells , Axons , Cell Differentiation
4.
Neural Regen Res ; 18(4): 746-749, 2023 Apr.
Article En | MEDLINE | ID: mdl-36204830

Neurite degeneration, a major component of many neurodegenerative diseases, such as Parkinson's disease, Alzheimer's disease, and amyotrophic lateral sclerosis, is not part of the typical apoptosis signaling mechanism, but rather it appears that a self-destructive process is in action. Oxidative stress is a well-known inducer of neurodegenerative pathways: neuronal cell death and neurite degeneration. Although oxidative stress exerts cytotoxic effects leading to neuronal loss, the pathogenic mechanisms and precise signaling pathways by which oxidative stress causes neurite degeneration have remained entirely unknown. We previously reported that reactive oxygen species generated by NADPH oxidases induce activation of the E3 ubiquitin ligase ZNRF1 in neurons, which promotes neurite degeneration. In this process, the phosphorylation of an NADPH oxidase subunit p47-phox at the 345th serine residue serves as an important checkpoint to initiate the ZNRF1-dependent neurite degeneration. Evidence provides new insights into the mechanism of reactive oxygen species-mediated neurodegeneration. In this review, we focus specifically on reactive oxygen species-induced neurite degeneration by highlighting a phosphorylation-dependent regulation of the molecular interaction between ZNRF1 and the NADPH oxidase complex.

5.
Exp Neurol ; 352: 114024, 2022 06.
Article En | MEDLINE | ID: mdl-35218706

Oxidative stress is a well-known inducer of two major neurodegenerative pathways, neuronal cell death and neurite degeneration. We previously reported that reactive oxygen species (ROS) generated by NADPH oxidases induces EGFR-dependent phosphorylation and activation of ZNRF1 ubiquitin ligase in neurons, which promotes neuronal cell death and neurite degeneration. While these findings provide a potential therapeutic avenue for neurodegeneration, a deeper understanding of the molecular mechanisms of this pathway have emerged as key points of interest. Here, we show that a NADPH oxidase subunit p47-phox/neutrophil cytosolic factor 1 regulates ZNRF1 activity. Using an in vitro neurite degeneration model, we demonstrate that transection-induced phosphorylation of p47-phox at the 345th serine residue by p38 MAPK serves as an initiating signal to activate ZNRF1. The phosphorylated p47 (pS345) or a phospho-mimetic mutant p47-phox binds directly to ZNRF1 whereas a phosphorylation-resistant mutant p47-phox cannot bind to ZNRF1 and its overexpression in neurites significantly suppresses ZNRF1 activation, AKT ubiquitination, and degeneration after transection, suggesting that pS345 might enhance the EGFR-mediated phosphorylation-dependent activation of ZNRF1. These results suggest that pS345 might represent an important checkpoint to initiate the ZNRF1-mediated neurite degeneration. Our findings provide novel insights into the mechanism of ROS-mediated neurodegeneration.


NADPH Oxidases , Serine , Enzyme Activation , ErbB Receptors/metabolism , NADPH Oxidases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Reactive Oxygen Species/metabolism , Serine/metabolism
6.
Front Mol Neurosci ; 14: 748721, 2021.
Article En | MEDLINE | ID: mdl-34630040

Small non-coding vault RNAs (vtRNAs) have been described as a component of the vault complex, a hollow-and-barrel-shaped ribonucleoprotein complex found in most eukaryotes. It has been suggested that the function of vtRNAs might not be limited to simply maintaining the structure of the vault complex. Despite the increasing research on vtRNAs, little is known about their physiological functions. Recently, we have shown that murine vtRNA (mvtRNA) up-regulates synaptogenesis by activating the mitogen activated protein kinase (MAPK) signaling pathway. mvtRNA binds to and activates mitogen activated protein kinase 1 (MEK1), and thereby enhances MEK1-mediated extracellular signal-regulated kinase activation. Here, we introduce the regulatory mechanism of MAPK signaling in synaptogenesis by vtRNAs and discuss the possibility as a novel molecular basis for synapse formation.

7.
Commun Integr Biol ; 14(1): 61-65, 2021 Apr 14.
Article En | MEDLINE | ID: mdl-33948134

The small non-coding vault RNA (vtRNA) is a component of the vault complex, a ribonucleoprotein complex found in most eukaryotes. vtRNAs regulate a variety of cellular functions when unassociated with the vault complex. Human has four vtRNA paralogs (hvtRNA1-1, hvtRNA1-2, hvtRNA1-3, hvtRNA2-1), which are highly similar and differ only slightly in primary and secondary structure. Despite the increasing research on vtRNAs, a feature that distinguishes one hvtRNA from the others has not been recognized. Recently, we demonstrated that murine vtRNA (mvtRNA) promotes synapse formation by modulating the MAPK signaling pathway. Here we showed that expression ofhvtRNA1-1, but not hvtRNA2-1 increases the expression of synaptic marker proteins, ERK phosphorylation and the number of PSD95 and Synapsin I double positive puncta to an extent similar to that of mvtRNA, suggesting that hvtRNA1-1 may enhance synapse formation. This finding opens new perspectives to uncover the function of the different vtRNA paralogs.

8.
J Cell Biol ; 220(2)2021 02 01.
Article En | MEDLINE | ID: mdl-33439240

The small noncoding vault RNA (vtRNA) is a component of the vault complex, a ribonucleoprotein complex found in most eukaryotes. Emerging evidence suggests that vtRNAs may be involved in the regulation of a variety of cellular functions when unassociated with the vault complex. Here, we demonstrate a novel role for vtRNA in synaptogenesis. Using an in vitro synapse formation model, we show that murine vtRNA (mvtRNA) promotes synapse formation by modulating the MAPK signaling pathway. mvtRNA is transported to the distal region of neurites as part of the vault complex. Interestingly, mvtRNA is released from the vault complex in the neurite by a mitotic kinase Aurora-A-dependent phosphorylation of MVP, a major protein component of the vault complex. mvtRNA binds to and activates MEK1 and thereby enhances MEK1-mediated ERK activation in neurites. These results suggest the existence of a regulatory mechanism of the MAPK signaling pathway by vtRNAs as a new molecular basis for synapse formation.


MAP Kinase Signaling System , RNA, Small Untranslated/metabolism , Synapses/metabolism , Amino Acid Sequence , Animals , Aurora Kinase A/metabolism , Cell Line , Down-Regulation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Kinesins/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred C57BL , Models, Biological , Neurites/metabolism , Oligonucleotides, Antisense/pharmacology , Post-Synaptic Density/drug effects , Post-Synaptic Density/metabolism , Protein Binding/drug effects , RNA, Small Interfering/metabolism , Synapses/drug effects , Vault Ribonucleoprotein Particles/chemistry , Vault Ribonucleoprotein Particles/metabolism
9.
J Clin Invest ; 131(1)2021 01 04.
Article En | MEDLINE | ID: mdl-33170806

Age-related sarcopenia constitutes an important health problem associated with adverse outcomes. Sarcopenia is closely associated with fat infiltration in muscle, which is attributable to interstitial mesenchymal progenitors. Mesenchymal progenitors are nonmyogenic in nature but are required for homeostatic muscle maintenance. However, the underlying mechanism of mesenchymal progenitor-dependent muscle maintenance is not clear, nor is the precise role of mesenchymal progenitors in sarcopenia. Here, we show that mice genetically engineered to specifically deplete mesenchymal progenitors exhibited phenotypes markedly similar to sarcopenia, including muscle weakness, myofiber atrophy, alterations of fiber types, and denervation at neuromuscular junctions. Through searching for genes responsible for mesenchymal progenitor-dependent muscle maintenance, we found that Bmp3b is specifically expressed in mesenchymal progenitors, whereas its expression level is significantly decreased during aging or adipogenic differentiation. The functional importance of BMP3B in maintaining myofiber mass as well as muscle-nerve interaction was demonstrated using knockout mice and cultured cells treated with BMP3B. Furthermore, the administration of recombinant BMP3B in aged mice reversed their sarcopenic phenotypes. These results reveal previously unrecognized mechanisms by which the mesenchymal progenitors ensure muscle integrity and suggest that age-related changes in mesenchymal progenitors have a considerable impact on the development of sarcopenia.


Aging/metabolism , Gene Expression Regulation , Growth Differentiation Factor 10/biosynthesis , Mesenchymal Stem Cells/metabolism , Muscle, Skeletal/metabolism , Sarcopenia/metabolism , Adult , Aging/genetics , Aging/pathology , Animals , Female , Growth Differentiation Factor 10/genetics , Humans , Male , Mesenchymal Stem Cells/pathology , Mice , Mice, Knockout , Middle Aged , Muscle, Skeletal/pathology , Sarcopenia/genetics , Sarcopenia/pathology
10.
Acta Neuropathol ; 140(5): 695-713, 2020 11.
Article En | MEDLINE | ID: mdl-32803350

Mislocalization and abnormal deposition of TDP-43 into the cytoplasm (TDP-43 proteinopathy) is a hallmark in neurons of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). However, the pathogenic mechanism of the diseases linked to TDP-43 is largely unknown. We hypothesized that the failure of mRNA transport to neuronal axons by TDP-43 may contribute to neurodegeneration in ALS and FTLD, and sought to examine the function of TDP-43 by identifying its target mRNA for axonal transport. We found that mRNAs related to translational function including ribosomal proteins (RPs) were decreased by shRNA-based TDP-43 knock-down in neurites of cortical neurons. TDP-43 binds to and transports the RP mRNAs through their 5' untranslated region, which contains a common 5' terminal oligopyrimidine tract motif and a downstream GC-rich region. We showed by employing in vitro and in vivo models that the RP mRNAs were translated and incorporated into native ribosomes locally in axons to maintain functionality of axonal ribosomes, which is required for local protein synthesis in response to stimulation and stress to axons. We also found that RP mRNAs were reduced in the pyramidal tract of sporadic ALS cases harboring TDP-43 pathology. Our results elucidated a novel function of TDP-43 to control transport of RP mRNAs and local translation by ribosomes to maintain morphological integrity of neuronal axons, and proved the influence of this function of TDP-43 on neurodegeneration in ALS and FTLD associated with TDP-43 proteinopathy.


DNA-Binding Proteins/metabolism , Protein Biosynthesis/physiology , Protein Transport/physiology , RNA, Messenger/metabolism , Ribosomal Proteins/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Axons/metabolism , Axons/pathology , Humans , Mice , Mice, Inbred C57BL , Neurons/metabolism , Neurons/pathology , TDP-43 Proteinopathies/metabolism , TDP-43 Proteinopathies/pathology
11.
J Neuroinflammation ; 16(1): 199, 2019 Oct 30.
Article En | MEDLINE | ID: mdl-31666085

BACKGROUND: Macrophage-derived high mobility group box 1 (HMGB1), a damage-associated molecular pattern (DAMP) protein, plays a key role in the development of chemotherapy-induced peripheral neuropathy (CIPN) caused by paclitaxel in rodents. Endothelial thrombomodulin (TM) promotes thrombin-induced degradation of HMGB1, and TMα, a recombinant human soluble TM, abolishes peripheral HMGB1-induced allodynia in mice. We thus examined whether HMGB1, particularly derived from macrophages, contributes to oxaliplatin-induced neuropathy in mice and analyzed the anti-neuropathic activity of the TM/thrombin system. METHODS: CIPN models were created by the administration of oxaliplatin in mice and rats, and the nociceptive threshold was assessed by von Frey test or paw pressure test. Macrophage-like RAW264.7 cells were stimulated with oxaliplatin in vitro. Proteins were detected and/or quantified by Western blotting, immunostaining, or enzyme-linked immunosorbent assay. RESULTS: Intraperitoneal administration of an anti-HMGB1-neutralizing antibody (AB) at 1 mg/kg prevented the oxaliplatin-induced allodynia in mice and rats. Antagonists of Toll-like receptor (TLR) 4, receptor for advanced glycation end products (RAGE) and CXCR4 among the HMGB1-targeted pro-nociceptive receptors, also mimicked the anti-neuropathic activity of AB in mice. Macrophage accumulation in the sciatic nerve was observed in mice treated with paclitaxel, but not oxaliplatin, and neither macrophage depletion nor inhibitors of macrophage activation affected oxaliplatin-induced allodynia. Oxaliplatin was 10- to 100-fold less potent than paclitaxel in releasing HMGB1 from macrophage-like RAW264.7 cells. Like AB, TMα at 10 mg/kg prevented the oxaliplatin-induced allodynia in mice as well as rats, an effect abolished by argatroban at 10 mg/kg, a thrombin inhibitor. The anti-neuropathic activity of TMα in oxaliplatin-treated mice was suppressed by oral anticoagulants such as warfarin at 1 mg/kg, dabigatran at 75 mg/kg, and rivaroxaban at 10 mg/kg, but not antiplatelet agents such as aspirin at 50 mg/kg and clopidogrel at 10 mg/kg. Repeated administration of the anticoagulants gradually developed neuropathic allodynia and elevated plasma HMGB1 levels in mice treated with a subeffective dose of oxaliplatin. CONCLUSIONS: Our data thus suggests a causative role of HMGB1 derived from non-macrophage cells in oxaliplatin-induced peripheral neuropathy and a thrombin-dependent anti-neuropathic activity of exogenous TMα and, most probably, endogenous TM.


Anticoagulants/administration & dosage , HMGB1 Protein/metabolism , Oxaliplatin/toxicity , Peripheral Nervous System Diseases/prevention & control , Thrombin/metabolism , Thrombomodulin/metabolism , Animals , Anticoagulants/adverse effects , Antineoplastic Agents/toxicity , Male , Mice , Peripheral Nervous System Diseases/chemically induced , RAW 264.7 Cells , Rats , Rats, Wistar , Rodentia
12.
Sci Rep ; 9(1): 7188, 2019 05 10.
Article En | MEDLINE | ID: mdl-31076621

Axonal degeneration occurs in various neurological diseases and traumatic nerve injury, and axonal regeneration is restricted by inhibitory factors in the central nervous system. Cyclin-dependent kinase 5 and glycogen synthase kinase 3ß (GSK3ß) are activated by one of those inhibitors, and collapsin response mediator protein 2 (CRMP2) is phosphorylated by both kinases. We previously developed a CRMP2 knock-in (CRMP2 KI) mouse line, in which CRMP2 phosphorylation at Ser 522 is inhibited. Because CRMP2 KI mice showed promotion of axonal regeneration after spinal cord injury, we hypothesized that CRMP2 KI mice would show higher axonal regeneration after optic nerve injury. In this study, we first show that depolymerization of microtubules after optic nerve crush (ONC) injury was suppressed in CRMP2 KI mice. Loss of retinal ganglia cells was also reduced after ONC. We found that protein level of GAP43, a marker of regenerative axons, was higher in the optic nerve from CRMP2KI than that from wild type 4 weeks after of ONC. We further observed increased numbers of axons labeled by tracer in the optic nerve after ONC in CRMP2 KI mice. These results suggest that inhibition of phosphorylation of CRMP2 suppresses axonal degeneration and promotes axonal regeneration after optic nerve injury.


Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Optic Nerve Injuries/therapy , Serine/metabolism , Animals , Disease Models, Animal , Gene Knock-In Techniques , Male , Mice , Microtubules , Nerve Crush , Nerve Regeneration , Optic Nerve Injuries/genetics , Optic Nerve Injuries/metabolism , Phosphorylation , Polymerization
13.
Biochem Biophys Res Commun ; 514(4): 1037-1039, 2019 07 05.
Article En | MEDLINE | ID: mdl-31097218

Axonal degeneration occurs in patients with various neurological diseases and traumatic nerve injuries, and Wallerian degeneration is a phenomenon in the prototypical axonal degradation that is observed after injury. Collapsin response mediator protein 2 (CRMP2) is phosphorylated by glycogen synthase kinase 3ß (GSK3ß), and it is involved in Wallerian degeneration after optic nerve injury. We previously developed a CRMP2 knock-in (CRMP2 KI) mouse line, in which CRMP2 phosphorylation by GSK3ß is inhibited; however, Wallerian degeneration in CRMP2 KI mice has not yet been examined. In this study, we examined whether Wallerian degeneration of the optic nerve is suppressed in CRMP2 KI mice. Using one eye removal model, we compared Wallerian degeneration of the optic nerve based on histological and biochemical analyses. Our experimental results indicated that the genetic inhibition of CRMP2 phosphorylation delays Wallerian degeneration after optic nerve injury.


Intercellular Signaling Peptides and Proteins/genetics , Nerve Tissue Proteins/genetics , Optic Nerve Injuries/genetics , Wallerian Degeneration/genetics , Animals , Disease Models, Animal , Intercellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Phosphorylation/drug effects , Phosphorylation/genetics , Semaphorin-3A/pharmacology
14.
Neurosci Res ; 139: 21-25, 2019 Feb.
Article En | MEDLINE | ID: mdl-30118738

ZNRF1 is an E3 ubiquitin ligase constitutively expressed in almost all neurons in the central and peripheral nervous systems during development and in adulthood. From this expression profile, the role of ZNRF1 is assumed to be common to many different types of neurons. We have analyzed the roles of ZNRF1-dependent degradation of target proteins in neurons. In mature neurons, ZNRF1 is activated in response to different types of stress that cause neuronal/axonal degeneration, and degrade AKT to activate GSK3B. Here we summarize the subcellular signaling events downstream of GSK3B activation, and their roles in the progression of neuronal/axonal degeneration.


Axons/metabolism , Carrier Proteins/metabolism , Neurons/metabolism , Ubiquitin/metabolism , Wallerian Degeneration/metabolism , Animals , Humans , Ubiquitin-Protein Ligases/metabolism , Wallerian Degeneration/pathology
15.
Neurosci Res ; 139: 58-62, 2019 Feb.
Article En | MEDLINE | ID: mdl-30194028

Pyridoxal, an active form of vitamin B6, is known to inhibit formation of advanced glycation end-products and protect tissues from diabetic complications. Here we identified that pyridoxal is a required component for establishing Schwann cell myelination in our Schwann cell-dorsal root ganglion neuron co-culture system. When the co-culture was maintained without pyridoxal, carboxymethylation of collapsin response mediator protein 2 (CRMP2) became detectable. Carboxymethylation decreased the affinity of CRMP2 to bind with microtubules, indicating that carboxymethylation affected CRMP2 function. These results suggest that carboxymethylation of CRMP2 may be an indicator of dysfunction caused by glycation which is observed in pathological conditions, including diabetic neuropathy.


Ganglia, Spinal/metabolism , Myelin Sheath/pathology , Neurons/metabolism , Schwann Cells/metabolism , Animals , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/pathology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Nerve Tissue Proteins/metabolism
16.
Neurosci Res ; 139: 63-68, 2019 Feb.
Article En | MEDLINE | ID: mdl-30194029

Amyotrophic lateral sclerosis (ALS) is an adult-onset neurological disease characterized by the selective degeneration of motor neurons leading to paralysis and immobility. Missense mutations in the gene coding for the Cu2+/Zn2+ superoxide dismutase 1 (SOD1) accounts for 15-20% of familial ALS, and mice overexpressing ALS-linked SOD1 mutants have been frequently used as an animal model for ALS. Degeneration of motor neurons in ALS progresses in a manner called "dying back", in which the degeneration of synapses and axons precedes the loss of cell bodies. Phosphorylation of collapsin response mediator protein 2 (CRMP2) is implicated in the progression of neuronal/axonal degeneration of different etiologies. To evaluate the role of CRMP2 phosphorylation in ALS pathogenesis, we utilized CRMP2 S522A knock-in (CRMP2ki/ki) mice, in which the serine residue 522 was homozygously replaced with alanine and thereby making CRMP2 no longer phosphorylatable by CDK5 or GSK3B. We found that the CRMP2ki/ki/SOD1G93A mice showed delay in the progression of the motor phenotype compared to their SOD1G93-Tg littermates. Histological analysis revealed that the CRMP2ki/ki/SOD1G93A mice retained more intact axons and NMJs than their SOD1G93A-Tg littermates. These results suggest that the phosphorylation of CRMP2 may contribute to the axonal degeneration of motor neurons in ALS.


Amyotrophic Lateral Sclerosis/metabolism , Axons/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Motor Neurons/metabolism , Nerve Tissue Proteins/metabolism , Amyotrophic Lateral Sclerosis/genetics , Animals , Disease Models, Animal , Mice, Transgenic , Neuromuscular Junction/pathology , Phosphorylation , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism , Synapses/metabolism
17.
Sci Rep ; 7(1): 5947, 2017 07 20.
Article En | MEDLINE | ID: mdl-28729624

Impairment of peripheral neurons by anti-cancer agents, including taxanes and platinum derivatives, has been considered to be a major cause of chemotherapy-induced peripheral neuropathy (CIPN), however, the precise underlying mechanisms are not fully understood. Here, we examined the direct effects of anti-cancer agents on Schwann cells. Exposure of primary cultured rat Schwann cells to paclitaxel (0.01 µM), cisplatin (1 µM), or oxaliplatin (3 µM) for 48 h induced cytotoxicity and reduced myelin basic protein expression at concentrations lower than those required to induce neurotoxicity in cultured rat dorsal root ganglion (DRG) neurons. Similarly, these anti-cancer drugs disrupted myelin formation in Schwann cell/DRG neuron co-cultures without affecting nerve axons. Cisplatin and oxaliplatin, but not paclitaxel, caused mitochondrial dysfunction in cultured Schwann cells. By contrast, paclitaxel led to dedifferentiation of Schwann cells into an immature state, characterized by increased expression of p75 and galectin-3. Consistent with in vitro findings, repeated injection of paclitaxel increased expression of p75 and galectin-3 in Schwann cells within the mouse sciatic nerve. These results suggest that taxanes and platinum derivatives impair Schwan cells by inducing dedifferentiation and mitochondrial dysfunction, respectively, which may be important in the development of CIPN in conjunction with their direct impairment in peripheral neurons.


Platinum/pharmacology , Schwann Cells/pathology , Taxoids/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Dedifferentiation/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Cells, Cultured , Cisplatin/pharmacology , Coculture Techniques , Galectin 3/metabolism , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Myelin Sheath/metabolism , Neurons/drug effects , Neurons/metabolism , Oxaliplatin/pharmacology , Paclitaxel/pharmacology , Rats , Receptor, Nerve Growth Factor/metabolism , Schwann Cells/drug effects , Schwann Cells/metabolism , Sciatic Nerve/cytology , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism
18.
Commun Integr Biol ; 10(2): e1296615, 2017.
Article En | MEDLINE | ID: mdl-28451058

Axonal degeneration is a key pathological feature of several neurological disorders. Emerging evidence has suggested a pathological connection between axonal degeneration and autophagy, a lysosomal degradation pathway. We recently reported that GSK3B-mediated phosphorylation of MCL1 regulates axonal autophagy to promote axonal degeneration. GSK3B-MCL1 pathway affects ATP production locally in degenerating axons and the exposure of phosphatidylserine (PS), an "eat-me" signal for phagocytes, on degenerating axons, resulting in the failed engulfment of axonal debris in vivo. Here we showed that the PS exposure is accomplished by phospholipid scramblase activity. This finding provides a novel mechanism that local ATP production through autophagy promotes PS exposure on degenerating axons. In addition, it opens new perspectives for the understanding of axonal autophagy to regulate Wallerian degeneration.

19.
J Cell Biol ; 216(2): 477-493, 2017 02.
Article En | MEDLINE | ID: mdl-28053206

Macroautophagy is a catabolic process, in which portions of cytoplasm or organelles are delivered to lysosomes for degradation. Emerging evidence has indicated a pathological connection between axonal degeneration and autophagy. However, the physiological function and induction mechanism of autophagy in axons remain elusive. We herein show that, through activation of BECLIN1, glycogen synthase kinase 3B (GSK3B)-mediated phosphorylation of BCL2 family member MCL1 induces axonal autophagy and axonal degeneration. Phosphorylated MCL1 is ubiquitinated by the FBXW7 ubiquitin ligase and degraded by the proteasome, thereby releasing BECLIN1 to induce axonal autophagy. Axonal autophagy contributes to local adenosine triphosphate production in degenerating axons and the exposure of phosphatidylserine-an "eat-me" signal for phagocytes-on transected axons and is required for normal recruitment of phagocytes to axonal debris in vivo. These results suggest that GSK3B-MCL1 signaling to regulate autophagy might be important for the successful completion of Wallerian degeneration.


Autophagy , Axons/enzymology , Ganglia, Spinal/enzymology , Glycogen Synthase Kinase 3 beta/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Wallerian Degeneration , Adenosine Triphosphate/metabolism , Animals , Axons/pathology , Beclin-1/metabolism , Cells, Cultured , F-Box Proteins/metabolism , F-Box-WD Repeat-Containing Protein 7 , Ganglia, Spinal/pathology , Glycogen Synthase Kinase 3 beta/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Phagocytes/metabolism , Phagocytosis , Phosphatidylserines/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proteolysis , RNA Interference , Signal Transduction , Time Factors , Transfection , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
20.
Sci Rep ; 6: 29856, 2016 07 19.
Article En | MEDLINE | ID: mdl-27432639

Rapid saltatory nerve conduction is facilitated by myelin structure, which is produced by Schwann cells (SC) in the peripheral nervous system (PNS). Proper development and degeneration/regeneration after injury requires regulated phenotypic changes of SC. We have previously shown that glutamate can induce SC proliferation in culture. Here we show that glutamate signals through metabotropic glutamate receptor 2 (mGluR2) to induce Erk phosphorylation in SC. mGluR2-elicited Erk phosphorylation requires ErbB2/3 receptor tyrosine kinase phosphorylation to limit the signaling cascade that promotes phosphorylation of Erk, but not Akt. We found that Gßγ and Src are involved in subcellular signaling downstream of mGluR2. We also found that glutamate can transform myelinating SC to proliferating SC, while inhibition of mGluR2 signaling can inhibit demyelination of injured nerves in vivo. These data suggest pathophysiological significance of mGluR2 signaling in PNS and its possible therapeutic importance to combat demyelinating disorders including Charcot-Marie-Tooth disease.


Glutamic Acid/genetics , Peripheral Nerve Injuries/genetics , Receptors, Metabotropic Glutamate/genetics , Schwann Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Glutamic Acid/metabolism , Humans , MAP Kinase Signaling System/genetics , Mice , Myelin Sheath/genetics , Myelin Sheath/metabolism , Peripheral Nerve Injuries/pathology , Peripheral Nervous System/injuries , Peripheral Nervous System/metabolism , Peripheral Nervous System/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Rats , Receptor, ErbB-2/genetics , Schwann Cells/pathology , Sciatic Nerve/injuries , Sciatic Nerve/pathology
...