Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
J Neuroinflammation ; 19(1): 179, 2022 Jul 12.
Article in English | MEDLINE | ID: mdl-35820932

ABSTRACT

BACKGROUND: Peripheral nerve injuries stimulate the regenerative capacity of injured neurons through a neuroimmune phenomenon termed the conditioning lesion (CL) response. This response depends on macrophage accumulation in affected dorsal root ganglia (DRGs) and peripheral nerves. The macrophage chemokine CCL2 is upregulated after injury and is allegedly required for stimulating macrophage recruitment and pro-regenerative signaling through its receptor, CCR2. In these tissues, CCL2 is putatively produced by neurons in the DRG and Schwann cells in the distal nerve. METHODS: Ccl2fl/fl mice were crossed with Advillin-Cre, P0-Cre, or both to create conditional Ccl2 knockouts (CKOs) in sensory neurons, Schwann cells, or both to hypothetically remove CCL2 and macrophages from DRGs, nerves or both. CCL2 was localized using Ccl2-RFPfl/fl mice. CCL2-CCR2 signaling was further examined using global Ccl2 KOs and Ccr2gfp knock-in/knock-outs. Unilateral sciatic nerve transection was used as the injury model, and at various timepoints, chemokine expression, macrophage accumulation and function, and in vivo regeneration were examined using qPCR, immunohistochemistry, and luxol fast blue staining. RESULTS: Surprisingly, in all CKOs, DRG Ccl2 gene expression was decreased, while nerve Ccl2 was not. CCL2-RFP reporter mice revealed CCL2 expression in several cell types beyond the expected neurons and Schwann cells. Furthermore, macrophage accumulation, myelin clearance, and in vivo regeneration were unaffected in all CKOs, suggesting CCL2 may not be necessary for the CL response. Indeed, Ccl2 global knockout mice showed normal macrophage accumulation, myelin clearance, and in vivo regeneration, indicating these responses do not require CCL2. CCR2 ligands, Ccl7 and Ccl12, were upregulated after nerve injury and perhaps could compensate for the absence of Ccl2. Finally, Ccr2gfp knock-in/knock-out animals were used to differentiate resident and recruited macrophages in the injured tissues. Ccr2gfp/gfp KOs showed a 50% decrease in macrophages in the distal nerve compared to controls with a relative increase in resident macrophages. In the DRG there was a small but insignificant decrease in macrophages. CONCLUSIONS: CCL2 is not necessary for macrophage accumulation, myelin clearance, and axon regeneration in the peripheral nervous system. Without CCL2, other CCR2 chemokines, resident macrophage proliferation, and CCR2-independent monocyte recruitment can compensate and allow for normal macrophage accumulation.


Subject(s)
Chemokine CCL2 , Macrophages , Peripheral Nerve Injuries , Animals , Axons/immunology , Axons/pathology , Chemokine CCL2/immunology , Chemokine CCL2/metabolism , Chemokines/immunology , Chemokines/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Nerve Regeneration/physiology , Peripheral Nerve Injuries/immunology , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/pathology
2.
Life Sci ; 287: 120117, 2021 Dec 15.
Article in English | MEDLINE | ID: mdl-34740577

ABSTRACT

Denervated skeletal muscular atrophy is primarily characterized by loss of muscle strength and mass and an unideal functional recovery of the muscle after extended denervation. This review emphasizes the interaction between the immune system and the denervated skeletal muscle. Immune cells such as neutrophils, macrophages and T-cells are activated and migrate to denervated muscle, where they release a high concentration of cytokines and chemokines. The migration of these immune cells, the transformation of different functional immune cell subtypes, and the cytokine network in the immune microenvironment may be involved in the regulatory process of muscle atrophy or repair. However, the exact mechanisms of the interaction between these immune cells and immune molecules in skeletal muscles are unclear. In this paper, the immune microenvironment regulation of muscle atrophy induced by peripheral nerve injury is reviewed.


Subject(s)
Biomedical Research/trends , Cellular Microenvironment/physiology , Immunity, Cellular/physiology , Muscle, Skeletal/immunology , Muscular Atrophy/immunology , Peripheral Nerve Injuries/immunology , Animals , Humans , Macrophages/immunology , Macrophages/metabolism , Muscle Denervation/methods , Muscle, Skeletal/innervation , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Peripheral Nerve Injuries/metabolism
3.
J Neuroinflammation ; 18(1): 227, 2021 Oct 13.
Article in English | MEDLINE | ID: mdl-34645458

ABSTRACT

BACKGROUND: Macrophages in the peripheral nervous system are key players in the repair of nerve tissue and the development of neuropathic pain due to peripheral nerve injury. However, there is a lack of information on the origin and morphological features of macrophages in sensory ganglia after peripheral nerve injury, unlike those in the brain and spinal cord. We analyzed the origin and morphological features of sensory ganglionic macrophages after nerve ligation or transection using wild-type mice and mice with bone-marrow cell transplants. METHODS: After protecting the head of C57BL/6J mice with lead caps, they were irradiated and transplanted with bone-marrow-derived cells from GFP transgenic mice. The infraorbital nerve of a branch of the trigeminal nerve of wild-type mice was ligated or the infraorbital nerve of GFP-positive bone-marrow-cell-transplanted mice was transected. After immunostaining the trigeminal ganglion, the structures of the ganglionic macrophages, neurons, and satellite glial cells were analyzed using two-dimensional or three-dimensional images. RESULTS: The number of damaged neurons in the trigeminal ganglion increased from day 1 after infraorbital nerve ligation. Ganglionic macrophages proliferated from days 3 to 5. Furthermore, the numbers of macrophages increased from days 3 to 15. Bone-marrow-derived macrophages increased on day 7 after the infraorbital nerve was transected in the trigeminal ganglion of GFP-positive bone-marrow-cell-transplanted mice but most of the ganglionic macrophages were composed of tissue-resident cells. On day 7 after infraorbital nerve ligation, ganglionic macrophages increased in volume, extended their processes between the neurons and satellite glial cells, and contacted these neurons. Most of the ganglionic macrophages showed an M2 phenotype when contact was observed, and little neuronal cell death occurred. CONCLUSION: Most of the macrophages that appear after a nerve injury are tissue-resident, and these make direct contact with damaged neurons that act in a tissue-protective manner in the M2 phenotype. These results imply that tissue-resident macrophages signal to neurons directly through physical contact.


Subject(s)
Bone Marrow Transplantation/methods , Cell Enlargement , Ganglia, Sensory/pathology , Macrophages/pathology , Peripheral Nerve Injuries/pathology , Sensory Receptor Cells/pathology , Animals , Ganglia, Sensory/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peripheral Nerve Injuries/immunology , Peripheral Nerve Injuries/therapy , Sensory Receptor Cells/immunology
4.
Acta Neuropathol Commun ; 9(1): 125, 2021 07 17.
Article in English | MEDLINE | ID: mdl-34274026

ABSTRACT

Peripheral nerve injury is a serious health problem and repairing long nerve deficits remains a clinical challenge nowadays. Nerve guidance conduit (NGC) serves as the most promising alternative therapy strategy to autografts but its repairing efficiency needs improvement. In this study, we investigated whether modulating the immune microenvironment by Interleukin-17F (IL-17F) could promote NGC mediated peripheral nerve repair. Chitosan conduits were used to bridge sciatic nerve defect in IL-17F knockout mice and wild-type mice with autografts as controls. Our data revealed that IL-17F knockout mice had improved functional recovery and axonal regeneration of sciatic nerve bridged by chitosan conduits comparing to the wild-type mice. Notably, IL-17F knockout mice had enhanced anti-inflammatory macrophages in the NGC repairing microenvironment. In vitro data revealed that IL-17F knockout peritoneal and bone marrow derived macrophages had increased anti-inflammatory markers after treatment with the extracts from chitosan conduits, while higher pro-inflammatory markers were detected in the Raw264.7 macrophage cell line, wild-type peritoneal and bone marrow derived macrophages after the same treatment. The biased anti-inflammatory phenotype of macrophages by IL-17F knockout probably contributed to the improved chitosan conduit guided sciatic nerve regeneration. Additionally, IL-17F could enhance pro-inflammatory factors production in Raw264.7 cells and wild-type peritoneal macrophages. Altogether, IL-17F may partially mediate chitosan conduit induced pro-inflammatory polarization of macrophages during nerve repair. These results not only revealed a role of IL-17F in macrophage function, but also provided a unique and promising target, IL-17F, to modulate the microenvironment and enhance the peripheral nerve regeneration.


Subject(s)
Chitosan , Guided Tissue Regeneration , Interleukin-17/genetics , Macrophages/immunology , Nerve Regeneration/immunology , Peripheral Nerve Injuries/immunology , Sciatic Nerve/physiology , Animals , Interleukin-17/immunology , Macrophages, Peritoneal/immunology , Mice , Mice, Knockout , Nerve Regeneration/physiology , RAW 264.7 Cells , Sciatic Nerve/surgery , Tissue Scaffolds
5.
Neurosci Lett ; 757: 135977, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34023413

ABSTRACT

BACKGROUND: Changes in inflammatory cytokine levels contribute to the induction and maintenance of neuropathic pain. We have shown that external low intensity focused ultrasound (liFUS) reduces allodynia in a common peroneal nerve injury (CPNI). Here, we investigate an underlying mechanism of action for this treatment and measure the effect of liFUS on inflammatory markers. METHODS: Male rats were divided into four groups: CPNI/liFUS, CPNI/shamliFUS, shamCPNI/liFUS, and shamCPNI/shamliFUS. Mechanical nociceptive thresholds were measured using Von Frey filaments (VFF) to confirm the absence/presence of allodynia at baseline, after CPNI, and after liFUS. Commercial microarray and ELISA assays were used to assess cytokine expression in the treated L5 dorsal root ganglion (DRG) and dorsal horn (DH) tissue 24 and 72 h after liFUS. RESULTS: VFF thresholds were significantly reduced following CPNI in both groups that received the injury (p < 0.001). After liFUS, only the CPNI/liFUS cohort showed a significant increase in mechanical thresholds (p < 0.001). CPNI significantly increased TNFa, IL6, CNTF, IL1b (p < 0.05 for all) levels in the DRG and DH, compared to baseline, consistent with previous work in sciatic nerve injury. LiFUS in CPNI rats resulted in a decrease in these cytokines in DRG 72 h post-therapy (TNFa, IL6, CNTF and IL1b, p < 0.001). In the DH, IL1b, CNTF, and TNFa (p < 0.05 for all) decreased 72 h after liFUS. CONCLUSION: We have demonstrated that liFUS modifies inflammatory cytokines in both DRG and DH in CPNI rats. These data provide evidence that liFUS, reverses the allodynic phenotype, in part, by altering inflammatory cytokine pathways.


Subject(s)
Hyperalgesia/therapy , Neuralgia/therapy , Peripheral Nerve Injuries/complications , Ultrasonic Therapy/methods , Animals , Cytokines/metabolism , Disease Models, Animal , Ganglia, Spinal/immunology , Ganglia, Spinal/metabolism , Humans , Hyperalgesia/diagnosis , Hyperalgesia/immunology , Male , Neuralgia/diagnosis , Neuralgia/immunology , Peripheral Nerve Injuries/immunology , Peripheral Nerve Injuries/therapy , Peroneal Nerve/injuries , Rats , Rats, Sprague-Dawley , Signal Transduction/immunology , Signal Transduction/radiation effects , Spinal Cord Dorsal Horn/immunology , Spinal Cord Dorsal Horn/metabolism , Ultrasonic Waves
6.
J Pain ; 22(10): 1146-1179, 2021 10.
Article in English | MEDLINE | ID: mdl-33892151

ABSTRACT

During persistent pain, the dorsal spinal cord responds to painful inputs from the site of injury, but the molecular modulatory processes have not been comprehensively examined. Using transcriptomics and multiplex in situ hybridization, we identified the most highly regulated receptors and signaling molecules in rat dorsal spinal cord in peripheral inflammatory and post-surgical incisional pain models. We examined a time course of the response including acute (2 hours) and longer term (2 day) time points after peripheral injury representing the early onset and instantiation of hyperalgesic processes. From this analysis, we identify a key population of superficial dorsal spinal cord neurons marked by somatotopic upregulation of the opioid neuropeptide precursor prodynorphin, and 2 receptors: the neurokinin 1 receptor, and anaplastic lymphoma kinase. These alterations occur specifically in the glutamatergic subpopulation of superficial dynorphinergic neurons. In addition to specific neuronal gene regulation, both models showed induction of broad transcriptional signatures for tissue remodeling, synaptic rearrangement, and immune signaling defined by complement and interferon induction. These signatures were predominantly induced ipsilateral to tissue injury, implying linkage to primary afferent drive. We present a comprehensive set of gene regulatory events across 2 models that can be targeted for the development of non-opioid analgesics. PERSPECTIVE: The deadly impact of the opioid crisis and the need to replace morphine and other opioids in clinical practice is well recognized. Embedded within this research is an overarching goal of obtaining foundational knowledge from transcriptomics to search for non-opioid analgesic targets. Developing such analgesics would address unmet clinical needs.


Subject(s)
Anaplastic Lymphoma Kinase/metabolism , Chronic Pain/metabolism , Hyperalgesia/metabolism , Neuroinflammatory Diseases/metabolism , Peripheral Nerve Injuries/metabolism , Posterior Horn Cells/metabolism , Transcriptome/physiology , Animals , Chronic Pain/immunology , Disease Models, Animal , Hyperalgesia/immunology , Neuroinflammatory Diseases/immunology , Peripheral Nerve Injuries/immunology , Posterior Horn Cells/immunology , Rats , Sequence Analysis, RNA
7.
J Dermatol Sci ; 102(2): 85-93, 2021 May.
Article in English | MEDLINE | ID: mdl-33676787

ABSTRACT

BACKGROUND: The involvement of the nerve in psoriasis development was suggested by sporadic case reports. OBJECTIVES: To provide multiple evidence for the nerve in psoriasis development with a retrospective case review, a literature review and a mouse-based experimental experiment. METHODS: Psoriatic patients who had concomitant nerve injuries and such cases from literatures were reviewed. And, on wild-type mouse level, unilateral denervation surgery was performed on the dorsal skin before and after the induction of psoriasiform dermatitis, respectively. Lesion visual scores were calculated, and biopsies were taken for hematoxylin-eosin (HE) staining, immunofluorescence analysis, and RNA sequencing & bioinformatics analysis before denervation surgery and the 2nd, 4th, 6th, 8th day after the surgery. RESULTS: All clinical cases (20/20) showed that local lesions under the control of injured nerves relieved spontaneously or even cleared/spared, and only about 1/3 experienced partial recurrence. Next, mouse psoriasiform experiments demonstrated that unilateral denervation prior to imiquimod application attenuated the enhancement of inflammatory reactions (e.g. adaptive immune response and Th17 cell differentiation pathway) and the induction of ipsilateral psoriasiform dermatitis. On the other hand, unilateral denervation after psoriasiform dermatitis induction promoted the regression of inflammatory reactions (e.g. T cell activation, TNF signaling, and Th17 cell differentiation pathway) and ipsilateral dermatitis recovery. CONCLUSION: Our study based on both retrospective clinical case review and wild-type mouse experiments provides multiple evidence for the involvement of the nerve in psoriasis development. Regulation of immune events, including TNF signaling and Th17 cell differentiation, may be the mechanisms of the nerve in psoriasis.


Subject(s)
Denervation , Neuroimmunomodulation , Peripheral Nerve Injuries/immunology , Psoriasis/surgery , Skin/innervation , Adult , Aged , Aged, 80 and over , Animals , Disease Models, Animal , Female , Gene Expression Regulation/immunology , Humans , Imiquimod/administration & dosage , Imiquimod/immunology , Male , Mice , Middle Aged , Peripheral Nerve Injuries/complications , Psoriasis/complications , Psoriasis/immunology , Retrospective Studies , Skin/immunology , Skin/pathology
8.
Neurorehabil Neural Repair ; 35(5): 431-443, 2021 05.
Article in English | MEDLINE | ID: mdl-33754913

ABSTRACT

Following traumatic peripheral nerve injury, adequate restoration of function remains an elusive clinical goal. Recent research highlights the complex role that the immune system plays in both nerve injury and regeneration. Pro-regenerative processes in wounded soft tissues appear to be significantly mediated by cytokines of the type 2 immune response, notably interleukin (IL)-4. While IL-4 signaling has been firmly established as a critical element in general tissue regeneration during wound healing, it has also emerged as a critical process in nerve injury and regeneration. In this context of peripheral nerve injury, endogenous IL-4 signaling has recently been confirmed to influence more than leukocytes, but including also neurons, axons, and Schwann cells. Given the role IL-4 plays in nerve injury and regeneration, exogenous IL-4 and/or compounds targeting this signaling pathway have shown encouraging preliminary results to treat nerve injury or other neuropathy in rodent models. In particular, the exogenous stimulation of the IL-4 signaling pathway appears to promote postinjury neuron survival, axonal regeneration, remyelination, and thereby improved functional recovery. These preclinical data strongly suggest that targeting IL-4 signaling pathways is a promising translational therapy to augment treatment approaches of traumatic nerve injury. However, a better understanding of the type 2 immune response and associated signaling networks functioning within the nerve injury microenvironment is still needed to fully develop this promising therapeutic avenue.


Subject(s)
Inflammation , Interleukin-4 , Nerve Regeneration/physiology , Peripheral Nerve Injuries , Signal Transduction/physiology , Animals , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Interleukin-4/immunology , Interleukin-4/metabolism , Nerve Regeneration/drug effects , Peripheral Nerve Injuries/drug therapy , Peripheral Nerve Injuries/immunology , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/physiopathology , Signal Transduction/drug effects
9.
Aging (Albany NY) ; 13(5): 6752-6764, 2021 02 17.
Article in English | MEDLINE | ID: mdl-33621204

ABSTRACT

Extracellular vesicles (EVs) are capable of transferring microRNAs (miRNAs or miRs) between two different types of cells and also serve as vehicles for delivery of therapeutic molecules. After peripheral nerve injury, abnormal expression patterns of miRNAs have been observed in dorsal root ganglia (DRG) sensory neurons. We hypothesized that sensory neurons secrete miRs-containing EVs to communicate with macrophages. We demonstrated that miR-23a was upregulated in DRG neurons in spared nerve injury (SNI) mouse models. We also found that miR-23a was enriched in EVs released by cultured DRG neurons following capsaicin treatment. miR-23a-containing EVs were taken up into macrophages in which increased intracellular miR-23a promoted pro-inflammatory phenotype. A20 was verified as a target gene of miR-23a. Moreover, intrathecal delivery of EVs-miR-23a antagomir attenuated neuropathic hypersensitivity and reduced the number of M1 macrophages in injured DRGs by targeting A20. In conclusion, these results demonstrate that sensory neurons transfer EVs-encapsulated miR-23a to activate M1 macrophages and enhance neuropathic pain following the peripheral nerve injury. The study highlighted a new therapeutic approach to alleviate chronic neuropathic pain after nerve trauma by targeting detrimental miRNA in sensory neurons.


Subject(s)
Extracellular Vesicles/metabolism , Macrophages/metabolism , Peripheral Nerve Injuries/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Animals , Disease Models, Animal , Extracellular Vesicles/genetics , Ganglia, Spinal/cytology , Mice , MicroRNAs/metabolism , Neuralgia/metabolism , Peripheral Nerve Injuries/immunology , Sensory Receptor Cells/metabolism , Up-Regulation
10.
Mol Cell Neurosci ; 111: 103590, 2021 03.
Article in English | MEDLINE | ID: mdl-33422671

ABSTRACT

Peripheral nerve injuries remain challenging to treat despite extensive research on reparative processes at the injury site. Recent studies have emphasized the importance of immune cells, particularly macrophages, in recovery from nerve injury. Macrophage plasticity enables numerous functions at the injury site. At early time points, macrophages perform inflammatory functions, but at later time points, they adopt pro-regenerative phenotypes to support nerve regeneration. Research has largely been limited, however, to the injury site. The neuromuscular junction (NMJ), the synapse between the nerve terminal and end target muscle, has received comparatively less attention, despite the importance of NMJ reinnervation for motor recovery. Macrophages are present at the NMJ following nerve injury. Moreover, in denervating diseases, such as amyotrophic lateral sclerosis (ALS), macrophages may also play beneficial roles at the NMJ. Evidence of positive macrophages roles at the injury site after peripheral nerve injury and at the NMJ in denervating pathologies suggest that macrophages may promote NMJ reinnervation. In this review, we discuss the intersection of nerve injury and immunity, with a focus on macrophages.


Subject(s)
Macrophages/immunology , Motor Neuron Disease/immunology , Neuromuscular Junction/immunology , Peripheral Nerve Injuries/immunology , Animals , Humans , Motor Neuron Disease/physiopathology , Nerve Regeneration , Neuromuscular Junction/physiology , Neuromuscular Junction/physiopathology , Peripheral Nerve Injuries/physiopathology
11.
Elife ; 92020 12 02.
Article in English | MEDLINE | ID: mdl-33263277

ABSTRACT

Sciatic nerve crush injury triggers sterile inflammation within the distal nerve and axotomized dorsal root ganglia (DRGs). Granulocytes and pro-inflammatory Ly6Chigh monocytes infiltrate the nerve first and rapidly give way to Ly6Cnegative inflammation-resolving macrophages. In axotomized DRGs, few hematogenous leukocytes are detected and resident macrophages acquire a ramified morphology. Single-cell RNA-sequencing of injured sciatic nerve identifies five macrophage subpopulations, repair Schwann cells, and mesenchymal precursor cells. Macrophages at the nerve crush site are molecularly distinct from macrophages associated with Wallerian degeneration. In the injured nerve, macrophages 'eat' apoptotic leukocytes, a process called efferocytosis, and thereby promote an anti-inflammatory milieu. Myeloid cells in the injured nerve, but not axotomized DRGs, strongly express receptors for the cytokine GM-CSF. In GM-CSF-deficient (Csf2-/-) mice, inflammation resolution is delayed and conditioning-lesion-induced regeneration of DRG neuron central axons is abolished. Thus, carefully orchestrated inflammation resolution in the nerve is required for conditioning-lesion-induced neurorepair.


Subject(s)
Ganglia, Spinal/immunology , Leukocytes/immunology , Macrophages/immunology , Nerve Regeneration , Peripheral Nerve Injuries/immunology , Phagocytosis , Sciatic Nerve/immunology , Animals , Apoptosis , Cells, Cultured , Cytokine Receptor Common beta Subunit/genetics , Cytokine Receptor Common beta Subunit/metabolism , Disease Models, Animal , Female , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Gene Expression Regulation , Gene Regulatory Networks , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Inflammation Mediators/metabolism , Leukocytes/metabolism , Leukocytes/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Neuronal Outgrowth , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/pathology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Signal Transduction
12.
Cells ; 9(9)2020 09 21.
Article in English | MEDLINE | ID: mdl-32967118

ABSTRACT

Microglia, the immunocompetent cells in the central nervous system (CNS), have long been studied as pathologically deteriorating players in various CNS diseases. However, microglia exert ameliorating neuroprotective effects, which prompted us to reconsider their roles in CNS and peripheral nervous system (PNS) pathophysiology. Moreover, recent findings showed that microglia play critical roles even in the healthy CNS. The microglial functions that normally contribute to the maintenance of homeostasis in the CNS are modified by other cells, such as astrocytes and infiltrated myeloid cells; thus, the microglial actions on neurons are extremely complex. For a deeper understanding of the pathophysiology of various diseases, including those of the PNS, it is important to understand microglial functioning. In this review, we discuss both the favorable and unfavorable roles of microglia in neuronal survival in various CNS and PNS disorders. We also discuss the roles of blood-borne macrophages in the pathogenesis of CNS and PNS injuries because they cooperatively modify the pathological processes of resident microglia. Finally, metabolic changes in glycolysis and oxidative phosphorylation, with special reference to the pro-/anti-inflammatory activation of microglia, are intensively addressed, because they are profoundly correlated with the generation of reactive oxygen species and changes in pro-/anti-inflammatory phenotypes.


Subject(s)
Cell Communication/immunology , Central Nervous System/immunology , Macrophages/immunology , Microglia/immunology , Nerve Regeneration/immunology , Peripheral Nervous System/immunology , Animals , Astrocytes/immunology , Astrocytes/metabolism , Astrocytes/pathology , Brain Infarction/immunology , Brain Infarction/metabolism , Brain Infarction/pathology , Brain Injuries, Traumatic/immunology , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Carbon Monoxide Poisoning/immunology , Carbon Monoxide Poisoning/metabolism , Carbon Monoxide Poisoning/pathology , Central Nervous System/metabolism , Central Nervous System/pathology , Glycolysis/genetics , Glycolysis/immunology , Humans , Macrophage Activation , Macrophages/metabolism , Macrophages/pathology , Microglia/metabolism , Microglia/pathology , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Oxidative Phosphorylation , Peripheral Nerve Injuries/immunology , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/pathology , Peripheral Nervous System/metabolism , Peripheral Nervous System/pathology , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism
13.
Methods Mol Biol ; 2143: 207-222, 2020.
Article in English | MEDLINE | ID: mdl-32524483

ABSTRACT

Injury to the sciatic nerve leads to degeneration and debris clearance in the area distal to the injury site, a process known as Wallerian degeneration. Immune cell infiltration into the distal sciatic nerve plays a major role in the degenerative process and subsequent regeneration of the injured motor and sensory axons. While macrophages have been implicated as the major phagocytic immune cell participating in Wallerian degeneration, recent work has found that neutrophils, a class of short-lived, fast responding white blood cells, also significantly contribute to the clearance of axonal and myelin debris. Detection of specific myeloid subtypes can be difficult as many cell-surface markers are often expressed on both neutrophils and monocytes/macrophages. Here we describe two methods for detecting neutrophils in the axotomized sciatic nerve of mice using immunohistochemistry and flow cytometry. For immunohistochemistry on fixed frozen tissue sections, myeloperoxidase and DAPI are used to specifically label neutrophils while a combination of Ly6G and CD11b are used to assess the neutrophil population of unfixed sciatic nerves using flow cytometry.


Subject(s)
Flow Cytometry/methods , Immunohistochemistry/methods , Neutrophils , Peripheral Nerve Injuries/pathology , Wallerian Degeneration/pathology , Animals , Antigens, Ly/analysis , Axotomy , Biomarkers , CD11b Antigen/analysis , Cell Separation , Fluorescent Dyes/analysis , Frozen Sections , Indoles/analysis , Mice , Neutrophils/enzymology , Neutrophils/pathology , Peripheral Nerve Injuries/immunology , Peroxidase/analysis , Phagocytosis , Sciatic Nerve/injuries , Sciatic Nerve/pathology , Staining and Labeling/methods , Wallerian Degeneration/immunology , von Willebrand Factor/analysis
14.
Eur J Pharmacol ; 880: 173171, 2020 Aug 05.
Article in English | MEDLINE | ID: mdl-32437743

ABSTRACT

Cathepsin S (CatS) is a cysteine protease found in lysosomes of hematopoietic and microglial cells and in secreted form in the extracellular space. While CatS has been shown to contribute significantly to neuropathic pain, the precise mechanisms remain unclear. In this report, we describe JNJ-39641160, a novel non-covalent, potent, selective and orally-available CatS inhibitor that is peripherally restricted (non-CNS penetrant) and may represent an innovative class of immunosuppressive and analgesic compounds and tools useful toward investigating peripheral mechanisms of CatS in neuropathic pain. In C57BL/6 mice, JNJ-39641160 dose-dependently blocked the proteolysis of the invariant chain, and inhibited both T-cell activation and antibody production to a vaccine antigen. In the spared nerve injury (SNI) model of chronic neuropathic pain, in which T-cell activation has previously been demonstrated to be a prerequisite for the development of pain hypersensitivity, JNJ-39641160 fully reversed tactile allodynia in wild-type mice but was completely ineffective in the same model in CatS knockout mice (which exhibited a delayed onset in allodynia). By contrast, in the acute mild thermal injury (MTI) model, JNJ-39641160 only weakly attenuated allodynia at the highest dose tested. These findings support the hypothesis that blockade of peripheral CatS alone is sufficient to fully reverse allodynia following peripheral nerve injury and suggest that the mechanism of action likely involves interruption of T-cell activation and peripheral cytokine release. In addition, they provide important insights toward the development of selective CatS inhibitors for the treatment of neuropathic pain in humans.


Subject(s)
Analgesics/therapeutic use , Cathepsins/antagonists & inhibitors , Hyperalgesia/drug therapy , Immunosuppressive Agents/therapeutic use , Neuralgia/drug therapy , Peripheral Nerve Injuries/drug therapy , Protease Inhibitors/therapeutic use , Analgesics/pharmacokinetics , Analgesics/pharmacology , Animals , Brain/metabolism , Cathepsins/genetics , Cathepsins/metabolism , Cell Line , Cytokines/immunology , Hot Temperature , Humans , Hyperalgesia/immunology , Immunoglobulin E/immunology , Immunoglobulin G/immunology , Immunosuppressive Agents/pharmacokinetics , Immunosuppressive Agents/pharmacology , Male , Mice, Inbred C57BL , Mice, Knockout , Neuralgia/immunology , Peripheral Nerve Injuries/immunology , Protease Inhibitors/pharmacokinetics , Protease Inhibitors/pharmacology , Sciatic Nerve/injuries , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tetanus Toxoid/administration & dosage , Touch
15.
Mol Immunol ; 121: 81-91, 2020 05.
Article in English | MEDLINE | ID: mdl-32172028

ABSTRACT

Traumatic injury to the peripheral nervous system (PNS) is the most common cause of acquired nerve damage and impairs the quality of life of patients. The success of nerve regeneration depends on distal stump degeneration, tissue clearance and remodeling, processes in which the immune system participates. We previously reported improved motor recovery in sciatic nerve crush mice following adoptive transfer of lymphocytes, which migrated to the lesion site. However, lymphocyte activity and the nerve tissue response remain unexplored. Thus, in the present study, we evaluated sciatic nerve regeneration and T cell polarization in lymphocyte recipient mice. Splenic lymphocytes were isolated from mice 14 days after sciatic nerve crush and transferred to axotomized animals three days postinjury. Immediate lymphocyte migration to the crushed nerve was confirmed by in vivo imaging. Phenotyping of T helper (Th) cells by flow cytometry revealed an increased frequency of the proinflammatory Th1 and Th17 cell subsets in recipient mice at 7 days and showed that the frequency of these cells remained unchanged for up to 21 days. Moreover, nerve regeneration was improved upon cell therapy, as shown by sustained immunolabeling of axons, Schwann cells, growth-associated protein 43 and BDNF from 14 to 28 days after lesion. Macrophage and IgG immunolabeling were also higher in cell-transferred mice at 14 and 21 days following nerve crush. Functionally, we observed better sensory recovery in the lymphocyte-treated group. Overall, our data demonstrate that enhanced inflammation early after nerve injury has beneficial effects for the regenerative process, improving tissue clearance and axonal regrowth towards the target organs.


Subject(s)
Adoptive Transfer/methods , Lymphocyte Transfusion , Nerve Regeneration/immunology , Peripheral Nerve Injuries/therapy , Sciatic Nerve/injuries , Animals , Axons/physiology , Disease Models, Animal , Humans , Male , Mice , Nerve Crush/adverse effects , Peripheral Nerve Injuries/immunology , Peripheral Nerve Injuries/pathology , Quality of Life , Sciatic Nerve/cytology , Sciatic Nerve/physiology , Spleen/cytology
16.
Eur J Pharmacol ; 872: 172949, 2020 Apr 05.
Article in English | MEDLINE | ID: mdl-31991141

ABSTRACT

Chronic pain conditions such as neuropathic pain and persistent inflammatory pain are difficult to manage. Alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors modulate nociceptive processing at the spinal dorsal horn. Previous studies have shown that intrathecal AMPA receptor antagonists exert antinociception in various pain states. Perampanel is a selective, noncompetitive inhibitor of the AMPA receptor and used clinically as an antiepileptic drug. Little is known about antinociceptive action of perampanel in the spinal cord. Here, we explored whether intrathecal perampanel attenuates neuropathic and inflammatory pain. A chronic constriction injury (CCI) to the sciatic nerve was induced in male Sprague-Dawley rats. We evaluated the effects of intrathecal perampanel (10, 30, or 100 µg) on mechanical and cold hyperalgesia using the electronic von Frey and cold plate tests, respectively. Normal rats were assessed in terms of inflammatory nociception using the formalin test, and motor function employing the rotarod test. In the CCI rats, spinally applied perampanel inhibited mechanical and cold hyperalgesia dose-dependently. In normal rats, perampanel remarkably suppressed the early- and late-phase responses in the formalin test, and it weakly affected motor performance for a short period at the highest dose. These results suggest that perampanel exerts antinociceptive actions on neuropathic and persistent inflammatory pain in the spinal cord. Perampanel may be safe and beneficial remedy for patients with such pain conditions. In addition, AMPA receptor can be a promising target for treatment of chronic pain.


Subject(s)
Chronic Pain/drug therapy , Neuralgia/drug therapy , Peripheral Nerve Injuries/drug therapy , Pyridones/administration & dosage , Receptors, AMPA/antagonists & inhibitors , Animals , Chronic Pain/immunology , Chronic Pain/pathology , Disease Models, Animal , Humans , Injections, Spinal , Male , Neuralgia/immunology , Neuralgia/pathology , Nitriles , Nociception/drug effects , Peripheral Nerve Injuries/complications , Peripheral Nerve Injuries/immunology , Rats , Sciatic Nerve/injuries , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/pathology
17.
Nat Commun ; 11(1): 264, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31937758

ABSTRACT

Paralleling the activation of dorsal horn microglia after peripheral nerve injury is a significant expansion and proliferation of macrophages around injured sensory neurons in dorsal root ganglia (DRG). Here we demonstrate a critical contribution of DRG macrophages, but not those at the nerve injury site, to both the initiation and maintenance of the mechanical hypersensitivity that characterizes the neuropathic pain phenotype. In contrast to the reported sexual dimorphism in the microglial contribution to neuropathic pain, depletion of DRG macrophages reduces nerve injury-induced mechanical hypersensitivity and expansion of DRG macrophages in both male and female mice. However, fewer macrophages are induced in the female mice and deletion of colony-stimulating factor 1 from sensory neurons, which prevents nerve injury-induced microglial activation and proliferation, only reduces macrophage expansion in male mice. Finally, we demonstrate molecular cross-talk between axotomized sensory neurons and macrophages, revealing potential peripheral DRG targets for neuropathic pain management.


Subject(s)
Ganglia, Spinal/immunology , Macrophages/physiology , Neuralgia/immunology , Animals , Cell Communication , Cell Proliferation/drug effects , Female , Hyperalgesia/immunology , Immunosuppressive Agents/pharmacology , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Microglia/metabolism , Microglia/physiology , Peripheral Nerve Injuries/immunology , Pregnancy , Sensory Receptor Cells/metabolism , Sex Factors , Tacrolimus/analogs & derivatives , Tacrolimus/pharmacology
19.
Brain Res Bull ; 155: 67-80, 2020 02.
Article in English | MEDLINE | ID: mdl-31756421

ABSTRACT

After peripheral axotomy, there is a selective retraction of synaptic terminals in contact with injured motoneurons. This process, which actively involves glial cells, is influenced by the expression of immune-related molecules. Since toll-like receptors (TLRs) are upregulated by astrocytes and microglia following lesions, they might be involved in synaptic plasticity processes. Therefore, we administered lipopolysaccharide (LPS) to enhance TLR4 expression in mice and studied retrograde changes in the spinal cord ventral horn following sciatic nerve crush. To this end, adult C57BL/6J male mice were subjected to unilateral sciatic nerve crush at the mid-thigh level and, after a survival time of seven and forty days (acute and chronic phases, respectively), the spinal cords were paraformaldehyde-fixed and dissected out for immunolabeling for synaptophysin, glial fibrillary acidic protein (GFAP) and ionized calcium-binding adapter molecule 1 (Iba1). The results show that TLR4 upregulation leads to synaptophysin downregulation close to spinal motoneuron cell bodies, indicating increased synaptic elimination. LPS exposure also further increases astrogliosis and microglial reactions in the both ventral and dorsal horns, especially ipsilateral to nerve axotomy, compared to those in untreated mice. Notably, LPS administration to TLR4-/- mice produces results similar to those observed in untreated wild-type counterparts, reinforcing the role of this receptor in the glial response to injury. Therefore, our results suggest that the overexpression of the TLR4 receptor results in augmented astrogliosis/microglial reactions and the excessive loss of synapses postinjury, which may, in turn, affect the motoneuronal regenerative response and functionality. Additionally, treatment with LPS increases the expression of ß2-microglobulin, a subcomponent of MHC I. Importantly, the absence of TLR4 results in imbalanced axonal regeneration, inducing subsequent improvements and setbacks. In conclusion, our results show the involvement of TLR4 in the process of synaptic remodeling, indicating a new target for future research aimed at developing therapies for CNS and PNS repair.


Subject(s)
Astrocytes/metabolism , Microglia/metabolism , Motor Neurons/metabolism , Peripheral Nerve Injuries/metabolism , Spinal Cord/metabolism , Toll-Like Receptor 4/metabolism , Animals , Lipopolysaccharides/administration & dosage , Male , Mice, Inbred C57BL , Nerve Crush , Neuronal Plasticity , Peripheral Nerve Injuries/immunology , Synaptophysin/metabolism
20.
Cell ; 176(4): 716-728.e18, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30712871

ABSTRACT

Sensory axons degenerate following separation from their cell body, but partial injury to peripheral nerves may leave the integrity of damaged axons preserved. We show that an endogenous ligand for the natural killer (NK) cell receptor NKG2D, Retinoic Acid Early 1 (RAE1), is re-expressed in adult dorsal root ganglion neurons following peripheral nerve injury, triggering selective degeneration of injured axons. Infiltration of cytotoxic NK cells into the sciatic nerve by extravasation occurs within 3 days following crush injury. Using a combination of genetic cell ablation and cytokine-antibody complex stimulation, we show that NK cell function correlates with loss of sensation due to degeneration of injured afferents and reduced incidence of post-injury hypersensitivity. This neuro-immune mechanism of selective NK cell-mediated degeneration of damaged but intact sensory axons complements Wallerian degeneration and suggests the therapeutic potential of modulating NK cell function to resolve painful neuropathy through the clearance of partially damaged nerves.


Subject(s)
Killer Cells, Natural/physiology , Nuclear Matrix-Associated Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Peripheral Nerve Injuries/metabolism , Animals , Axons , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Killer Cells, Natural/metabolism , Male , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Nerve Regeneration , Neurons/cytology , Neurons, Afferent/immunology , Neurons, Afferent/metabolism , Nuclear Matrix-Associated Proteins/physiology , Nucleocytoplasmic Transport Proteins/physiology , Pain , Peripheral Nerve Injuries/immunology , Peripheral Nervous System Diseases , Sciatic Nerve , Sensory Receptor Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...