Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 269
Filter
1.
Muscle Nerve ; 70(3): 420-433, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38922958

ABSTRACT

INTRODUCTION/AIMS: The precise relationship between molecular mimicry and tissue-specific autoimmunity is unknown. Major histocompatibility complex (MHC) class II antigen presenting cell-CD4+ T-cell receptor complex interactions are necessary for adaptive immunity. This study aimed to determine the role of endoneurial endothelial cell MHC class II in autoimmune polyneuropathy. METHODS: Cryopreserved Guillain-Barré syndrome (GBS) patient sural nerve biopsies and sciatic nerves from the severe murine experimental autoimmune neuritis (sm-EAN) GBS model were studied. Cultured conditional ready MHC Class II antigen A-alpha chain (H2-Aa) embryonic stem cells were used to generate H2-Aaflox/+ C57BL/6 mice. Mice were backcrossed and intercrossed to the SJL background to generate H2-Aaflox/flox SJL mice, bred with hemizygous Tamoxifen-inducible von Willebrand factor Cre recombinase (vWF-iCre/+) SJL mice to generate H2-Aaflox/flox; vWF-iCre/+ mice to study microvascular endothelial cell adaptive immune responses. Sm-EAN was induced in Tamoxifen-treated H2-Aaflox/flox; vWF-iCre/+, H2-Aaflox/flox; +/+, H2-Aa+/+; vWF-iCre/+ and untreated H2-Aaflox/flox; vWF-iCre/+ adult female SJL mice. Neurobehavioral, electrophysiological and histopathological assessments were performed at predefined time points. RESULTS: Endoneurial endothelial cell MHC class II expression was observed in normal and inflamed human and mouse peripheral nerves. Tamoxifen-treated H2-Aaflox/flox; vWF-iCre/+ mice were resistant to sm-EAN despite extensive MHC class II expression in lymphoid and non-lymphoid tissues. DISCUSSION: A conditional MHC class II knockout mouse to study cell- and time-dependent adaptive immune responses in vivo was developed. Initial studies show microvascular endothelial cell MHC class II expression is necessary for peripheral nerve specific autoimmunity, as advocated by human in vitro adaptive immunity and ex vivo transplant rejection studies.


Subject(s)
Adaptive Immunity , Histocompatibility Antigens Class II , Mice, Inbred C57BL , Mice, Knockout , Animals , Mice , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/genetics , Humans , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/genetics , Guillain-Barre Syndrome/pathology , Neuritis, Autoimmune, Experimental/immunology , Neuritis, Autoimmune, Experimental/pathology , Neuritis, Autoimmune, Experimental/genetics , Sciatic Nerve/immunology , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Peripheral Nerves/immunology , Peripheral Nerves/metabolism , Peripheral Nerves/pathology , Female , Endothelial Cells/immunology , Endothelial Cells/metabolism , Sural Nerve/pathology , Sural Nerve/immunology , Male , Time Factors
2.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35058362

ABSTRACT

Immune cells infiltrate the peripheral nervous system (PNS) after injury and with autoimmunity, but their net effect is divergent. After injury, immune cells are reparative, while in inflammatory neuropathies (e.g., Guillain Barré Syndrome and chronic inflammatory demyelinating polyneuropathy), immune cells are proinflammatory and promote autoimmune demyelination. An understanding of immune cell phenotypes that distinguish these conditions may, therefore, reveal new therapeutic targets for switching immune cells from an inflammatory role to a reparative state. In an autoimmune regulator (Aire)-deficient mouse model of inflammatory neuropathy, we used single-cell RNA sequencing of sciatic nerves to discover a transcriptionally heterogeneous cellular landscape, including multiple myeloid, innate lymphoid, and lymphoid cell types. Analysis of cell-cell ligand-receptor interactions uncovered a macrophage-mediated tumor necrosis factor-α (TNF-α) signaling axis that is induced by interferon-γ and required for initiation of autoimmune demyelination. Developmental trajectory visualization suggested that TNF-α signaling is associated with metabolic reprogramming of macrophages and polarization of macrophages from a reparative state in injury to a pathogenic, inflammatory state in autoimmunity. Autocrine TNF-α signaling induced macrophage expression of multiple genes (Clec4e, Marcksl1, Cxcl1, and Cxcl10) important in immune cell activation and recruitment. Genetic and antibody-based blockade of TNF-α/TNF-α signaling ameliorated clinical neuropathy, peripheral nerve infiltration, and demyelination, which provides preclinical evidence that the TNF-α axis may be effectively targeted to resolve inflammatory neuropathies.


Subject(s)
Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Peripheral Nervous System Diseases/etiology , Peripheral Nervous System Diseases/metabolism , Polyendocrinopathies, Autoimmune/complications , Tumor Necrosis Factor-alpha/metabolism , Adoptive Transfer , Animals , Antibodies, Monoclonal/pharmacology , Autocrine Communication , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Transgenic , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/pathology , Paracrine Communication , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/pathology , Polyendocrinopathies, Autoimmune/genetics , Receptors, Tumor Necrosis Factor/deficiency , Sciatic Nerve/immunology , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors
3.
J Tissue Eng Regen Med ; 15(11): 1023-1036, 2021 11.
Article in English | MEDLINE | ID: mdl-34591344

ABSTRACT

Decellularized extra-cellular matrix (ECM) has been studied as an alternative to anti-adhesive biomaterials and cartilage acellular matrix (CAM) has been shown to inhibit postoperative adhesion in several organs. This study aimed to evaluate the suitability of glutaraldehyde (GA) crosslinked CAM-films as anti-adhesion barriers for peripheral nerve injury. The films were successfully fabricated and showed improved physical properties such as mechanical strength, swelling ratio, and lengthened degradation period while maintaining the microstructure and chemical composition after GA crosslinking. In the in vitro study of CAM-film, the dsDNA content met the recommended limit of decellularization and more than 70% of the major ECM components were preserved after decellularization. The adhesion and proliferation of seeded human umbilical vein endothelial cells and fibroblasts were significantly lower in CAM-film than in control, but similar with Seprafilm. However, the CAM-film extract did not show cytotoxicity. In the in vivo study, the peri-neural fibrosis was thicker, adhesion score higher, and peri-neural collagen fibers more abundant in the control group than in the CAM-film group. The total number of myelinated axons was significantly higher in the CAM-film group than in the control group. The inflammatory marker decreased with time in the CAM-film group compared to that in the control group, whereas the nerve regenerative marker expression was maintained. Moreover, the ankle angles at contracture and toe-off were higher in the CAM film-treated rats than in the control rats. GA-crosslinked CAM films may be used during peripheral nerve surgery to prevent peri-neural adhesion and enhance nerve functional recovery.


Subject(s)
Cartilage/chemistry , Cross-Linking Reagents/chemistry , Extracellular Matrix/chemistry , Glutaral/chemistry , Nerve Regeneration/physiology , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Animals , Cell Adhesion , Cell Death , Cell Proliferation , Collagen/metabolism , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice , Rats, Sprague-Dawley , Recovery of Function/drug effects , Sciatic Nerve/immunology , Sciatic Nerve/pathology , Swine
4.
Front Immunol ; 12: 720733, 2021.
Article in English | MEDLINE | ID: mdl-34484228

ABSTRACT

One hallmark of Guillain-Barre syndrome (GBS), a prototypic autoimmune peripheral neuropathy (APN) is infiltration of leukocytes (macrophages and T cells) into peripheral nerves, where chemokines and their receptors play major roles. In this study, we aimed to understand the potential contribution of chemokine receptors CCR2 and CX3CR1 in APN by using a well-established mouse model, B7.2 transgenic (L31) mice, which possesses a predisposed inflammatory background. We crossbred respectively CCR2KO and CX3CR1KO mice with L31 mice. The disease was initiated by partial ligation on one of the sciatic nerves. APN pathology and neurological function were evaluated on the other non-ligated sciatic nerve/limb. Our results revealed that L31/CX3CR1KO but not L31/CCR2KO mice were resistant to APN. CX3CR1 is needed for maintaining circulating monocyte and CD8+ T cell survival. While migration of a significant number of activated CD8+ T cells to peripheral nerves is essential in autoimmune response in nerve, recruitment of monocytes into PNS seems optional. Disease onset is independent of CCR2 mediated blood-derived macrophage recruitment, which can be replaced by compensatory proliferation of resident macrophages in peripheral nerve. CX3CR1 could also contribute to APN via its critical involvement in maintaining nerve macrophage phagocytic ability. We conclude that blockade of CX3CR1 signaling may represent an interesting anti-inflammatory strategy to improve therapeutic management for GBS patients.


Subject(s)
Autoimmunity/genetics , CX3C Chemokine Receptor 1/genetics , Gene Expression , Neuritis, Autoimmune, Experimental/etiology , Peripheral Nervous System Diseases/etiology , Receptors, CCR2/genetics , Animals , Biomarkers , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CX3C Chemokine Receptor 1/metabolism , Disease Models, Animal , Disease Susceptibility/immunology , Immunophenotyping , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Neuritis, Autoimmune, Experimental/metabolism , Neuritis, Autoimmune, Experimental/pathology , Peripheral Nervous System Diseases/metabolism , Receptors, CCR2/metabolism , Sciatic Nerve/immunology , Sciatic Nerve/metabolism , Sciatic Nerve/pathology
5.
Int Immunopharmacol ; 99: 107918, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34320458

ABSTRACT

OBJECT: Ninjurin2 (nerve injury induced protein 2, NINJ2) is a molecule which mediates cell-to-cell and cell-to-extracellular matrix interactions in the nervous system. Clinical study shows NINJ2 is associated with the development of postherpetic neuralgia. However, it is lack of direct evidence that NINJ2 participated in neuropathic pain. In this study, we aim to investigate the role of NINJ2 in the development of neuropathic pain in spared sciatic nerve injury rats and the underlying mechanism. METHOD: Spared sciatic nerve injury (SNI) models were established. The level of NINJ2 and p-p65 (a NF-κB family member) were measured in SNI rats by western blots and immunofluorescent staining. Lentivirus encoding small interfering RNA targeting NINJ2 (RNAi) was intrathecally injected into rats. Then the change of pain behavior of rats induced by NINJ2 RNAi was tested by Von-Frey hairs. The change of p-p65 in the spinal cord in rats after NINJ2 RNAi treatment was also measured by western blots. inhibitor of p-p65-induced change of TNF-α, IL-1ß, and IL-6 levels were measured by ELISA. RESULTS: NINJ2 and p-p65 were increased in the spinal cord of SNI rats on the 3, 7, 14th days after modeling. NINJ2 were mainly expressed in neurons, and co-located with p-p65 in the spinal dorsal horn. When down regulating the level of NINJ2 by RNAi, the development of pain in SNI rats was partially blocked. Phosphorylation of p65 was also inhibited by NINJ2 RNAi. Blocking the phosphorylation of NF-κB pathway could inhibit the increase of TNF-α, IL-1ß, and IL-6 in the spinal cord of SNI rats. CONCLUSION: NINJ2 protein was increased in the spinal cord of SNI rats. It participated in the development of nerve injury-induced neuropathic pain by activating neuroinflammation in the spinal cord via NF-κB pathway. This study provides a new target to investigate the mechanism of neuropathic pain.


Subject(s)
Cell Adhesion Molecules, Neuronal/immunology , Neuralgia/immunology , Neuroinflammatory Diseases/immunology , Sciatic Nerve/injuries , Transcription Factor RelA/immunology , Animals , Cell Adhesion Molecules, Neuronal/genetics , Male , Rats, Sprague-Dawley , Sciatic Nerve/immunology , Spinal Cord/immunology
6.
Front Immunol ; 12: 650269, 2021.
Article in English | MEDLINE | ID: mdl-34093538

ABSTRACT

Inflammation is a hallmark of several neurodegenerative disorders including hereditary amyloidogenic transthyretin amyloidosis (ATTRv). ATTRv is an autosomal dominant neurodegenerative disorder with extracellular deposition of mutant transthyretin (TTR) aggregates and fibrils, particularly in nerves and ganglia of the peripheral nervous system. Nerve biopsies from ATTRv patients show increased cytokine production, but interestingly no immune inflammatory cellular infiltrate is observed around TTR aggregates. Here we show that as compared to Wild Type (WT) animals, the expression of several chemokines is highly downregulated in the peripheral nervous system of a mouse model of the disease. Interestingly, we found that stimulation of mouse Schwann cells (SCs) with WT TTR results in the secretion of several chemokines, a process that is mediated by toll-like receptor 4 (TLR4). In contrast, the secretion of all tested chemokines is compromised upon stimulation of SCs with mutant TTR (V30M), suggesting that V30M TTR fails to activate TLR4 signaling. Altogether, our data shed light into a previously unappreciated mechanism linking TTR activation of SCs and possibly underlying the lack of inflammatory response observed in the peripheral nervous system of ATTRv patients.


Subject(s)
Amyloid Neuropathies, Familial/immunology , Chemokines/metabolism , Down-Regulation/immunology , Prealbumin/genetics , Amyloid Neuropathies, Familial/genetics , Amyloid Neuropathies, Familial/pathology , Animals , Cells, Cultured , Disease Models, Animal , Humans , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Transgenic , Mutation , Prealbumin/isolation & purification , Prealbumin/metabolism , Primary Cell Culture , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Schwann Cells/immunology , Schwann Cells/metabolism , Schwann Cells/pathology , Sciatic Nerve/immunology , Sciatic Nerve/pathology , Toll-Like Receptor 4/metabolism
7.
J Exp Med ; 218(8)2021 08 02.
Article in English | MEDLINE | ID: mdl-34086056

ABSTRACT

Peripheral nerve injury can cause debilitating disease and immune cell-mediated destruction of the affected nerve. While the focus has been on the nerve-regenerative response, the effect of loss of innervation on lymph node function is unclear. Here, we show that the popliteal lymph node (popLN) receives direct neural input from the sciatic nerve and that sciatic denervation causes lymph node expansion. Loss of sympathetic, adrenergic tone induces the expression of IFN-γ in LN CD8 T cells, which is responsible for LN expansion. Surgery-induced IFN-γ expression and expansion can be rescued by ß2 adrenergic receptor agonists but not sensory nerve agonists. These data demonstrate the mechanisms governing the pro-inflammatory effect of loss of direct adrenergic input on lymph node function.


Subject(s)
Adrenergic Agents/metabolism , Interferon-gamma/metabolism , Lymph Nodes/pathology , Peripheral Nerve Injuries/pathology , Animals , Antigens/immunology , Autoimmunity , Axotomy , CD8-Positive T-Lymphocytes/immunology , Denervation , Inflammation/pathology , Male , Mice, Inbred C57BL , Sciatic Nerve/immunology , Sciatic Nerve/pathology , Signal Transduction
8.
Peptides ; 136: 170447, 2021 02.
Article in English | MEDLINE | ID: mdl-33212101

ABSTRACT

The selection of control group is crucial, as the use of an inadequate group may strongly affect the results. In this study we examine the effect on contralateral tissue protein levels, in a model of unilateral UVB irradiation, as the contralateral side is commonly used as a control. Previous studies have shown that UVB irradiation increases immunoreactivity for inflammatory regulated neuropeptides. Unilateral UVB irradiation of rat hind paw was performed and corresponding contralateral spinal cord and dorsal root ganglia (DRG) were collected 2-96 h after and investigated for changes in galanin, substance P and c-fos immunoreactivity. Control tissue was collected from naïve rats. Measurement of skin blood flow from contralateral heel hind paws (Doppler), revealed no change compared to naïve rats. However, UVB irradiation caused a significant reduction in the contralateral proportion of galanin immunopositive DRG neurons, at all-time points, as well as an increase in the contralateral spinal cord dorsal horn, around the central canal and in the lateral spinal nucleus (2-48 h). The contralateral proportion of SP positive DRG neurons and dorsal horn immunoreactivity was unchanged, whereas the lateral spinal nucleus area showed increased immunoreactivity (48 h). UVB irradiation also induced a slight contralateral upregulation of c-fos in the dorsal horn/central canal area (24 and 48 h). In summary, unilateral UVB irradiation induced contralateral changes in inflammatory/nociceptive neuropeptides in spinal cord and afferent pathways involved in pain signaling already within 24 h, a time point when also ipsilateral neurochemical/physiological changes have been reported for rats and humans.


Subject(s)
Galanin/immunology , Neurons/immunology , Proto-Oncogene Proteins c-fos/immunology , Substance P/immunology , Animals , Galanin/radiation effects , Ganglia, Spinal/immunology , Ganglia, Spinal/radiation effects , Humans , Medulla Oblongata/immunology , Medulla Oblongata/radiation effects , Neurons/radiation effects , Neuropeptides/genetics , Pain/immunology , Pain/pathology , Proto-Oncogene Proteins c-fos/radiation effects , Rats , Sciatic Nerve/immunology , Sciatic Nerve/radiation effects , Spinal Cord/immunology , Spinal Cord/radiation effects , Spinal Cord Dorsal Horn/metabolism , Spinal Cord Dorsal Horn/radiation effects , Substance P/radiation effects , Ultraviolet Rays/adverse effects
9.
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
10.
Front Immunol ; 11: 575792, 2020.
Article in English | MEDLINE | ID: mdl-33329541

ABSTRACT

Recent preclinical and clinical evidence suggest that immune system has a role in the progression and prognosis of Amyotrophic Lateral Sclerosis (ALS), but the identification of a clear mechanism and immune players remains to be elucidated. Here, we have investigated, in 30 and 60 days (presymptomatic) and 120 days (symptomatic) old SOD1-G93A mice, systemic, peripheral, and central innate and adaptive immune and inflammatory response, correlating it with the progression of the neurodegeneration in neuromuscular junction, sciatic nerves, and spinal cord. Surprisingly, we found a very initial (45-60 days) presence of IgG in sciatic nerves together with a gradual enhancement of A20/TNFAIP3 (protein controlling NF-κB signalling) and a concomitantly significant increase and activation of circulating mast cells (MCs) as well as MCs and macrophages in sciatic nerve and an enhancement of IL-6 and IL-10. This immunological frame coincided with a myelin aggregation. The 30-60 days old SOD1-G93A mice didn't show real elements of neuroinflammation and neurodegeneration in spinal cord. In 120 days old mice macrophages and monocytes are widely diffused in sciatic nerves, peripheral neurodegeneration reaches the tip, high circulating levels of TNFα and IL-2 were found and spinal cord exhibits clear signs of neural damage and infiltrating immune cells. Our results underpin a clear immunological disorder at the origin of ALS axonopathy, in which MCs are involved in the initiation and sustaining of inflammatory events. These data cannot be considered a mere epiphenomenon of motor neuron degeneration and reveal new potential selective immune targets in ALS therapy.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Immunity, Innate , Neuroimmunomodulation , Neuromuscular Junction/immunology , Sciatic Nerve/immunology , Spinal Cord/immunology , Superoxide Dismutase-1/metabolism , Wallerian Degeneration , Amyotrophic Lateral Sclerosis/enzymology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Disease Progression , Genetic Predisposition to Disease , Interleukin-10/metabolism , Interleukin-6/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Mast Cells/immunology , Mast Cells/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Mutation , NF-kappa B/metabolism , Neuromuscular Junction/enzymology , Neuromuscular Junction/pathology , Phenotype , Sciatic Nerve/enzymology , Sciatic Nerve/pathology , Signal Transduction , Spinal Cord/enzymology , Spinal Cord/pathology , Superoxide Dismutase-1/genetics , Time Factors
11.
Nat Neurosci ; 23(11): 1339-1351, 2020 11.
Article in English | MEDLINE | ID: mdl-33077946

ABSTRACT

Microglia and peripheral macrophages have both been implicated in amyotrophic lateral sclerosis (ALS), although their respective roles have yet to be determined. We now show that macrophages along peripheral motor neuron axons in mouse models and patients with ALS react to neurodegeneration. In ALS mice, peripheral myeloid cell infiltration into the spinal cord was limited and depended on disease duration. Targeted gene modulation of the reactive oxygen species pathway in peripheral myeloid cells of ALS mice, using cell replacement, reduced both peripheral macrophage and microglial activation, delayed symptoms and increased survival. Transcriptomics revealed that sciatic nerve macrophages and microglia reacted differently to neurodegeneration, with abrupt temporal changes in macrophages and progressive, unidirectional activation in microglia. Modifying peripheral macrophages suppressed proinflammatory microglial responses, with a shift toward neuronal support. Thus, modifying macrophages at the periphery has the capacity to influence disease progression and may be of therapeutic value for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/immunology , Axons/immunology , Macrophages/immunology , Microglia/immunology , Motor Neurons/immunology , Sciatic Nerve/immunology , Adult , Aged , Amyotrophic Lateral Sclerosis/metabolism , Animals , Female , Humans , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Middle Aged , Motor Neurons/metabolism , Sciatic Nerve/metabolism , Spinal Cord/immunology , Spinal Cord/metabolism
12.
J Neurosci Res ; 98(12): 2468-2495, 2020 12.
Article in English | MEDLINE | ID: mdl-32931034

ABSTRACT

Ablation/segmental loss peripheral nerve injuries (PNIs) exhibit poor functional recovery due to slow and inaccurate outgrowth of regenerating axons. Viable peripheral nerve allografts (PNAs) as growth-guide conduits are immunologically rejected and all anucleated donor/host axonal segments undergo Wallerian degeneration. In contrast, we report that ablation-type sciatic PNIs repaired by neurorrhaphy of viable sciatic PNAs and a polyethylene glycol (PEG)-fusion protocol using PEG immediately restored axonal continuity for many axons, reinnervated/maintained their neuromuscular junctions, and prevented much Wallerian degeneration. PEG-fused PNAs permanently restored many sciatic-mediated behaviors within 2-6 weeks. PEG-fused PNAs were not rejected even though host/donors were neither immunosuppressed nor tissue-matched in outbred female Sprague Dawley rats. Innate and adaptive immune responses to PEG-fused sciatic PNAs were analyzed using electron microscopy, immunohistochemistry, and quantitative reverse transcription polymerase chain reaction for morphological features, T cell and macrophage infiltration, major histocompatibility complex (MHC) expression, apoptosis, expression of cytokines, chemokines, and cytotoxic effectors. PEG-fused PNAs exhibited attenuated innate and adaptive immune responses by 14-21 days postoperatively, as evidenced by (a) many axons and cells remaining viable, (b) significantly reduced infiltration of cytotoxic and total T cells and macrophages, (c) significantly reduced expression of inflammatory cytokines, chemokines, and MHC proteins, (d) consistently low apoptotic response. Morphologically and/or biochemically, PEG-fused sciatic PNAs often resembled sciatic autografts or intact sciatic nerves. In brief, PEG-fused PNAs are an unstudied, perhaps unique, example of immune tolerance of viable allograft tissue in a nonimmune-privileged environment and could greatly improve the clinical outcomes for PNIs relative to current protocols.


Subject(s)
Adaptive Immunity/physiology , Immunity, Innate/physiology , Polyethylene Glycols/administration & dosage , Sciatic Nerve/immunology , Sciatic Nerve/transplantation , Sciatic Neuropathy/therapy , Adaptive Immunity/drug effects , Allografts/immunology , Allografts/transplantation , Animals , Female , Immunity, Innate/drug effects , Male , Rats , Rats, Sprague-Dawley , Sciatic Neuropathy/immunology , Transplantation, Homologous/methods
13.
Int J Mol Sci ; 21(17)2020 Aug 31.
Article in English | MEDLINE | ID: mdl-32878186

ABSTRACT

The acceleration of peripheral nerve regeneration is crucial for functional nerve recovery. Our previous study demonstrated that human Wharton's jelly-derived mesenchymal stem cells (hWJ-MSC) promote sciatic nerve recovery and regeneration via the direct upregulation and release of neurotrophic factors. However, the immunomodulatory role of hWJ-MSC in sciatic nerve recovery remains unclear. The effects of hWJ-MSC on innate immunity, represented by macrophages, natural killer cells, and dendritic cells, as well as on adaptive immunity, represented by CD4+ T, CD8+ T, B, and regulatory T cells (Tregs), were examined using flow cytometry. Interestingly, a significantly increased level of Tregs was detected in blood, lymph nodes (LNs), and nerve-infiltrating cells on POD7, 15, 21, and 35. Anti-inflammatory cytokines, such as IL-4 and IL-10, were significantly upregulated in the LNs and nerves of hWJ-MSC-treated mice. Treg depletion neutralized the improved effects of hWJ-MSC on sciatic nerve recovery. In contrast, Treg administration promoted the functional recovery of five-toe spread and gait stance. hWJ-MSC also expressed high levels of the anti-inflammatory cytokines TGF-ß and IL-35. This study indicated that hWJ-MSC induce Treg development to modulate the balance between pro- and anti-inflammation at the injured sciatic nerve by secreting higher levels of anti-inflammatory cytokines.


Subject(s)
Cytokines/metabolism , Mesenchymal Stem Cells/cytology , Sciatic Nerve/cytology , T-Lymphocytes, Regulatory/immunology , Wharton Jelly/cytology , Animals , Cell Proliferation , Cells, Cultured , Immunologic Factors/metabolism , Mesenchymal Stem Cells/immunology , Mice , Mice, Inbred BALB C , Sciatic Nerve/immunology , Wharton Jelly/immunology
14.
J Exp Med ; 217(11)2020 11 02.
Article in English | MEDLINE | ID: mdl-32648893

ABSTRACT

Wallerian degeneration (WD) is a process of autonomous distal degeneration of axons upon injury. Macrophages (MPs) of the peripheral nervous system (PNS) are the main cellular agent controlling this process. Some evidence suggests that resident PNS-MPs along with MPs of hematogenous origin may be involved, but whether these two subsets exert distinct functions is unknown. Combining MP-designed fluorescent reporter mice and coherent anti-Stokes Raman scattering (CARS) imaging of the sciatic nerve, we deciphered the spatiotemporal choreography of resident and recently recruited MPs after injury and unveiled distinct functions of these subsets, with recruited MPs being responsible for efficient myelin stripping and clearance and resident MPs being involved in axonal regrowth. This work provides clues to tackle selectively cellular processes involved in neurodegenerative diseases.


Subject(s)
Macrophages/immunology , Wallerian Degeneration/diagnostic imaging , Wallerian Degeneration/immunology , Animals , Axons/physiology , Disease Models, Animal , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Sheath/physiology , Nonlinear Optical Microscopy , Remyelination/genetics , Sciatic Nerve/diagnostic imaging , Sciatic Nerve/immunology , Sciatic Nerve/injuries , Transcriptome
15.
J Vis Exp ; (160)2020 06 14.
Article in English | MEDLINE | ID: mdl-32597850

ABSTRACT

This protocol describes a footpad inoculation model used to study the initiation and development of neuroinflammatory responses during alphaherpesvirus infection in mice. As alphaherpesviruses are main invaders of the peripheral nervous system (PNS), this model is suitable to characterize the kinetics of viral replication, its spread from the PNS to CNS, and associated neuroinflammatory responses. The footpad inoculation model allows virus particles to spread from a primary infection site in the footpad epidermis to sensory and sympathetic nerve fibers that innervate the epidermis, sweat glands, and dermis. The infection spreads via the sciatic nerve to the dorsal root ganglia (DRG) and ultimately through the spinal cord to the brain. Here, a mouse footpad is inoculated with pseudorabies virus (PRV), an alphaherpesvirus closely related to herpes simplex virus (HSV) and varicella-zoster virus (VZV). This model demonstrates that PRV infection induces severe inflammation, characterized by neutrophil infiltration in the footpad and DRG. High concentrations of inflammatory cytokines are subsequently detected in homogenized tissues by ELISA. In addition, a strong correlation is observed between PRV gene and protein expression (via qPCR and IF staining) in DRG and the production of pro-inflammatory cytokines. Therefore, the footpad inoculation model provides a better understanding of the processes underlying alphaherpesvirus-induced neuropathies and may lead to the development of innovative therapeutic strategies. In addition, the model can guide research on peripheral neuropathies, such as multiple sclerosis and associated viral-induced damage to the PNS. Ultimately, it can serve as a cost-effective in vivo tool for drug development.


Subject(s)
Alphaherpesvirinae/immunology , Ganglia, Spinal/immunology , Herpesviridae Infections/immunology , Hindlimb/virology , Inflammation/etiology , Peripheral Nervous System Diseases/etiology , Sciatic Nerve/immunology , Animals , Disease Models, Animal , Ganglia, Spinal/virology , Herpesviridae Infections/complications , Herpesviridae Infections/virology , Inflammation/pathology , Mice , Mice, Inbred C57BL , Peripheral Nervous System Diseases/pathology , Sciatic Nerve/virology , Virus Replication
16.
Nat Neurosci ; 23(5): 676-689, 2020 05.
Article in English | MEDLINE | ID: mdl-32284604

ABSTRACT

While CNS microglia have been extensively studied, relatively little is known about macrophages populating the peripheral nervous system. Here we performed ontogenic, transcriptomic and spatial characterization of sciatic nerve macrophages (snMacs). Using multiple fate-mapping systems, we show that snMacs do not derive from the early embryonic precursors colonizing the CNS, but originate primarily from late embryonic precursors and become replaced by bone-marrow-derived macrophages over time. Using single-cell transcriptomics, we identified a tissue-specific core signature of snMacs and two spatially separated snMacs: Relmα+Mgl1+ snMacs in the epineurium and Relmα-Mgl1- snMacs in the endoneurium. Globally, snMacs lack most of the core signature genes of microglia, with only the endoneurial subset expressing a restricted number of these genes. In response to nerve injury, the two resident snMac populations respond differently. Moreover, and unlike in the CNS, monocyte-derived macrophages that develop during injury can engraft efficiently in the pool of resident peripheral nervous system macrophages.


Subject(s)
Macrophages/cytology , Macrophages/physiology , Sciatic Nerve/immunology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Nerve Crush , Transcriptome
17.
Int J Neurosci ; 130(11): 1109-1117, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32009498

ABSTRACT

Background: Guillain-Barré syndrome (GBS), an autoimmune disease and an acute inflammation disorder, is currently the most frequent cause of acute flaccid paralysis worldwide. EAN, an animal model of GBS, is a CD4+ T cell-mediated autoimmune disease of the PNS. Wnt/ß-catenin signals are critically important to several fundamental aspects of peripheral nerve development and play a crucial role in Schwann cell proliferation. Here, we investigate the role of Wnt/ß-catenin signalling cascades in EAN rats.Methods: 28 male Lewis rats weighing 170 ± 10 g were randomly divided into control group (n = 7) and EAN groups (Early group; Peak group and Recovery group. n = 7 per group). EAN rats were immunized with P257-81 peptide; weighed daily, and the neurologic signs of EAN were evaluated every day. The sciatic nerve was taken on the days 10, 17, and 30 p.i. for H&E staining, transmission electron microscopy and immunohistochemical staining; blood samples were collected weekly from caudal vein to detect IFN-γ, IL-4, TGF-ß1; and the sciatic nerve was taken to examinate the dynamics expression of Wnt/ß-catenin pathway molecules.Results: In our study, we chose tail-root injection to better model GBS. Moreover, we observed that IFN-γ levels paralleled clinical EAN, and the levels of TGF-ß1 and IL-4 gradually increased and peaked in the recovery phase. In addition, we have shown that canonical Wnt signalling is upregulated and reached a peak in the late recovery phase.Conclusion: Our findings suggest that Wnt/ß-catenin signalling is associated with the promotion of remyelination in EAN rats.


Subject(s)
Guillain-Barre Syndrome , Interferon-gamma/blood , Interleukin-4/blood , Neuritis, Autoimmune, Experimental , Remyelination , Sciatic Nerve , Transforming Growth Factor beta1/blood , Wnt Signaling Pathway , Animals , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/metabolism , Guillain-Barre Syndrome/pathology , Male , Neuritis, Autoimmune, Experimental/immunology , Neuritis, Autoimmune, Experimental/metabolism , Neuritis, Autoimmune, Experimental/pathology , Rats , Rats, Inbred Lew , Remyelination/physiology , Sciatic Nerve/immunology , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Up-Regulation , Wnt Signaling Pathway/physiology
18.
Neurotherapeutics ; 17(1): 235-242, 2020 01.
Article in English | MEDLINE | ID: mdl-31673865

ABSTRACT

Our study objective was testing for anti-neuronal autoantibodies within commercially available intravenous immunoglobulin (IVIg) preparations. Sixteen samples from 5 different commercially available IVIg preparations were tested with cell-based assays (CBA) and enzyme-linked immunosorbent assay (ELISA) to detect and characterize common neuronal autoantibodies, and with immunohistochemistry on teased fibers from mouse sciatic nerve and on mouse brain sections to screen for nodal and not yet identified neuronal antigens. In 15/16 IVIg preparations, anti-GAD antibodies were detected in titers ranging from 40 to 1507 IU/mL, as typically seen in type 1 diabetes, but not in the range (> 2000 IU/mL) seen in GAD-positive neurological patients. None of the preparations was however positive with anti-GAD CBA. Antibodies to AQP4 were also detected by ELISA in 15/16 IVIg preparations with titers comparable to those seen in AQP4-seropositive NMO patients; with CBA, however, all IVIg samples were AQP4-negative. IVIg preparations contained IgG-anti-MAG antibodies by ELISA at statistically significant higher titers compared to controls. Two of the 16 IVIg samples were positive for human 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) antibodies. All IVIg preparations were negative for antibodies to MOG, NMDAR, anti-nodal, and other neuronal-specific proteins. IVIg preparations contain antibodies against GAD and AQP4 in titers comparable to those seen in autoimmune patients when tested by ELISA, but not by CBA or tissue immunohistochemistry, suggesting that the autoantibodies within the IVIg are against linear rather than structural epitopes, as part of the natural antibody immune repertoire. The information is clinically important for diagnosis when testing patients' sera after they have received therapy with IVIg to avoid false interpretation.


Subject(s)
Autoantibodies/analysis , Immunoglobulins, Intravenous/analysis , Neurons/immunology , Animals , Aquaporin 4/analysis , Aquaporin 4/immunology , Autoantibodies/immunology , Enzyme-Linked Immunosorbent Assay , Immunoglobulins, Intravenous/immunology , Mice , Sciatic Nerve/immunology
19.
Int. j. morphol ; 37(4): 1527-1533, Dec. 2019. tab, graf
Article in Spanish | LILACS | ID: biblio-1040165

ABSTRACT

En órganos dañados, el ácido láctico (AL) modifica la respuesta inmune innata e inflamatoria, induciendo una menor expresión de citoquinas pro-inflamatorias, que provocan, la modulación del reclutamiento de células inmunes. El daño por compresión del nervio isquiático (NI) desencadena una respuesta inflamatoria y un aumento exponencial del infiltrado inflamatorio de células inmunes, produciendo la destrucción de axones y pérdida funcional del nervio. El objetivo de este estudio es evaluar el efecto agudo de la inyección de AL, sobre la proporción de células inmunes en la fase inflamatoria temprana, en el sitio de lesión del NI post compresión. Para ello, se utilizaron 15 ratas machos Sprague Dawley adultas, en tres grupos de compresión nerviosa. Un grupo control, un grupo control negativo con placebo (100 µL PBS) y un grupo experimental con inyección de 100 µL de AL [20mM]. Al tercer día los NI se analizaron histológicamente y se estableció la proporción de células inmunes en el sitio de lesión. Los resultados muestran que la inyección intraneural de AL provoca una disminución en el porcentaje de linfocitos y un aumento en el porcentaje de macrófagos. Este es el primer trabajo de inyección intraneural de AL y demuestra el efecto modulador del AL sobre las células inmunes en el sistema nervioso periférico.


In damaged organs, lactic acid (LA) modifies the innate and inflammatory immune response, inducing a lower expression of pro-inflammatory cytokines, which provoke the modulation of immune cell recruitment. Damage by compression of the sciatic nerve (SN) triggers an inflammatory response and an exponential increase in the inflammatory infiltrate of immune cells, producing the destruction of axons and functional loss of the nerve. The objective of this study is to evaluate the acute effect of the injection of LA, on the proportion of immune cells in the early inflammatory phase, in the site of SN post-compression injury. For this, 15 adult Sprague Dawley rats were used in three groups of nervous compression. A control group, a negative control group with placebo (100 mL PBS) and an experimental group with injection of 100 mL of LA [20mM]. On the third day, the SNs were histologically analyzed and the proportion of immune cells at the injury site was established. The results show that the intraneural injection of LA causes a decrease in the percentage of lymphocytes and an increase in the percentage of macrophages. This is the first work of intraneural injection of LA and demonstrates the modulating effect of LA on immune cells in the peripheral nervous system.


Subject(s)
Animals , Male , Rats , Sciatic Nerve/drug effects , Sciatic Nerve/immunology , Lactic Acid/pharmacology , Nerve Compression Syndromes/pathology , Sciatic Nerve/pathology , Lymphocytes/drug effects , Cytokines/immunology , Cytokines/metabolism , Rats, Sprague-Dawley , Lactic Acid/administration & dosage , Inflammation/immunology , Macrophages/drug effects
20.
Front Immunol ; 10: 2132, 2019.
Article in English | MEDLINE | ID: mdl-31552056

ABSTRACT

Objective: Dimethyl fumarate (DMF) exerts immunomodulatory and neuroprotective effects in the animal model of experimental autoimmune neuritis (EAN) in the Lewis rat. DMF has been shown to modulate gut microbiota in veterinary medicine, however the effects of oral DMF on the gut-associated lymphoid tissue (GALT) remain unknown. Methods: Lewis rats were treated orally twice daily with DMF up to day 10 after immunization with immunogenic P2 peptide. Histological, flow cytometric and RT-PCR analyses of the GALT (intraepithelial layer, lamina propria, and Peyer patches) in duodenum, jejunum, and ileum were performed ex vivo. Moreover, cell transfer experiments were used to examine the protective effects of GALT regulatory T cells of the Peyer patches. Results: In the upper layers of duodenum, DMF induced a reduction of the toll-like receptor 4 (TLR4) mRNA expression. This was combined by a decrease of the pro-inflammatory lamina propria IFN-γ mRNA expression. In the ileum, we detected an immunoregulatory phenotype characterized by an increase of FoxP3 mRNA expression and of the nuclear factor (erythroid-derived-2)- like 2 (Nrf2) downstream molecule heme oxygenase-1 (HO-1) mRNA. Finally, CD4+ CD25+ regulatory T cells were increased in the Peyer patches. In vivo, the protective effect of these regulatory cells was verified by cell transfer into recipient EAN rats. Conclusions: Our results identified a novel immunomodulatory effect of DMF through the different regions and layers of the small intestine, which led to an increase of regulatory T cells, exerting a protective role in experimental neuritis.


Subject(s)
Dimethyl Fumarate/therapeutic use , Immunologic Factors/therapeutic use , Intestine, Small/drug effects , Neuritis, Autoimmune, Experimental/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Cytokines/genetics , Dimethyl Fumarate/pharmacology , Female , Immunologic Factors/pharmacology , Intestine, Small/immunology , Neuritis, Autoimmune, Experimental/immunology , Neuroprotective Agents/pharmacology , Peyer's Patches/drug effects , Peyer's Patches/immunology , Rats, Inbred Lew , Sciatic Nerve/drug effects , Sciatic Nerve/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology
SELECTION OF CITATIONS
SEARCH DETAIL