69 research outputs found

    Acute- and late-phase matrix metalloproteinase (MMP)-9 activity is comparable in female and male rats after peripheral nerve injury.

    Get PDF
    BACKGROUND:In the peripheral nerve, pro-inflammatory matrix metalloproteinase (MMP)-9 performs essential functions in the acute response to injury. Whether MMP-9 activity contributes to late-phase injury or whether MMP-9 expression or activity after nerve injury is sexually dimorphic remains unknown. METHODS:Patterns of MMP-9 expression, activity and excretion were assessed in a model of painful peripheral neuropathy, sciatic nerve chronic constriction injury (CCI), in female and male rats. Real-time Taqman RT-PCR for MMP-9 and its endogenous inhibitor, tissue inhibitor of metalloproteinase-1 (TIMP-1) of nerve samples over a 2-month time course of CCI was followed by gelatin zymography of crude nerve extracts and purified MMP-9 from the extracts using gelatin Sepharose-beads. MMP excretion was determined using protease activity assay of urine in female and male rats with CCI. RESULTS:The initial upsurge in nerve MMP-9 expression at day 1 post-CCI was superseded more than 100-fold at day 28 post-CCI. The high level of MMP-9 expression in late-phase nerve injury was accompanied by the reduction in TIMP-1 level. The absence of MMP-9 in the normal nerve and the presence of multiple MMP-9 species (the proenzyme, mature enzyme, homodimers, and heterodimers) was observed at day 1 and day 28 post-CCI. The MMP-9 proenzyme and mature enzyme species dominated in the early- and late-phase nerve injury, consistent with the high and low level of TIMP-1 expression, respectively. The elevated nerve MMP-9 levels corresponded to the elevated urinary MMP excretion post-CCI. All of these findings were comparable in female and male rodents. CONCLUSION:The present study offers the first evidence for the excessive, uninhibited proteolytic MMP-9 activity during late-phase painful peripheral neuropathy and suggests that the pattern of MMP-9 expression, activity, and excretion after peripheral nerve injury is universal in both sexes

    The alternatively spliced fibronectin CS1 isoform regulates IL-17A levels and mechanical allodynia after peripheral nerve injury.

    Get PDF
    BackgroundMechanical pain hypersensitivity associated with physical trauma to peripheral nerve depends on T-helper (Th) cells expressing the algesic cytokine, interleukin (IL)-17A. Fibronectin (FN) isoform alternatively spliced within the IIICS region encoding the 25-residue-long connecting segment 1 (CS1) regulates T cell recruitment to the sites of inflammation. Herein, we analyzed the role of CS1-containing FN (FN-CS1) in IL-17A expression and pain after peripheral nerve damage.MethodsMass spectrometry, immunoblotting, and FN-CS1-specific immunofluorescence analyses were employed to examine FN expression after chronic constriction injury (CCI) in rat sciatic nerves. The acute intra-sciatic nerve injection of the synthetic CS1 peptide (a competitive inhibitor of the FN-CS1/Ξ±4 integrin binding) was used to elucidate the functional significance of FN-CS1 in mechanical and thermal pain hypersensitivity and IL-17A expression (by quantitative Taqman RT-PCR) after CCI. The CS1 peptide effects were analyzed in cultured primary Schwann cells, the major source of FN-CS1 in CCI nerves.ResultsFollowing CCI, FN expression in sciatic nerve increased with the dominant FN-CS1 deposition in endothelial cells, Schwann cells, and macrophages. Acute CS1 therapy attenuated mechanical allodynia (pain from innocuous stimulation) but not thermal hyperalgesia and reduced the levels of IL-17A expression in the injured nerve. CS1 peptide inhibited the LPS- or starvation-stimulated activation of the stress ERK/MAPK pathway in cultured Schwann cells.ConclusionsAfter physical trauma to the peripheral nerve, FN-CS1 contributes to mechanical pain hypersensitivity by increasing the number of IL-17A-expressing (presumably, Th17) cells. CS1 peptide therapy can be developed for pharmacological control of neuropathic pain

    Obesity Takes Its Toll on Visceral Pain: High-Fat Diet Induces Toll-Like Receptor 4- Dependent Visceral Hypersensitivity

    Get PDF
    Exposure to high-fat diet induces both, peripheral and central alterations in TLR4 expression. Moreover, functional TLR4 is required for the development of high-fat diet-induced obesity. Recently, central alterations in TLR4 expression have been associated with the modulation of visceral pain. However, it remains unknown whether there is a functional interaction between the role of TLR4 in diet-induced obesity and in visceral pain. In the present study we investigated the impact of long-term exposure to high-fat diet on visceral pain perception and on the levels of TLR4 and Cd11b (a microglial cell marker) protein expression in the prefrontal cortex (PFC) and hippocampus. Peripheral alterations in TLR4 were assessed following the stimulation of spleenocytes with the TLR4-agonist LPS. Finally, we evaluated the effect of blocking TLR4 on visceral nociception, by administering TAK-242, a selective TLR4-antagonist. Our results demonstrated that exposure to high-fat diet induced visceral hypersensitivity. In parallel, enhanced TLR4 expression and microglia activation were found in brain areas related to visceral pain, the PFC and the hippocampus. Likewise, peripheral TLR4 activity was increased following long-term exposure to high-fat diet, resulting in an increased level of pro-inflammatory cytokines. Finally, TLR4 blockage counteracted the hyperalgesic phenotype present in mice fed on high-fat diet. Our data reveal a role for TLR4 in visceral pain modulation in a model of diet-induced obesity, and point to TLR4 as a potential therapeutic target for the development of drugs to treat visceral hypersensitivity present in pathologies associated to fat diet consumption

    Immunodominant fragments of myelin basic protein initiate T cell-dependent pain

    Get PDF
    AbstractBackgroundThe myelin sheath provides electrical insulation of mechanosensory AΞ²-afferent fibers. Myelin-degrading matrix metalloproteinases (MMPs) damage the myelin sheath. The resulting electrical instability of AΞ²-fibers is believed to activate the nociceptive circuitry in AΞ²-fibers and initiate pain from innocuous tactile stimulation (mechanical allodynia). The precise molecular mechanisms, responsible for the development of this neuropathic pain state after nerve injury (for example, chronic constriction injury, CCI), are not well understood.Methods and resultsUsing mass spectrometry of the whole sciatic nerve proteome followed by bioinformatics analyses, we determined that the pathways, which are classified as the Infectious Disease and T-helper cell signaling, are readily activated in the nerves post-CCI. Inhibition of MMP-9/MMP-2 suppressed CCI-induced mechanical allodynia and concomitant TNF-Ξ± and IL-17A expression in nerves. MMP-9 proteolysis of myelin basic protein (MBP) generated the MBP84-104 and MBP68-86 digest peptides, which are prominent immunogenic epitopes. In agreement, the endogenous MBP69-86 epitope co-localized with MHCII and MMP-9 in Schwann cells and along the nodes of Ranvier. Administration of either the MBP84-104 or MBP68-86 peptides into the naΓ―ve nerve rapidly produced robust mechanical allodynia with a concomitant increase in T cells and MHCII-reactive cell populations at the injection site. As shown by the genome-wide expression profiling, a single intraneural MBP84-104 injection stimulated the inflammatory, immune cell trafficking, and antigen presentation pathways in the injected naΓ―ve nerves and the associated spinal cords. Both MBP84-104-induced mechanical allodynia and characteristic pathway activation were remarkably less prominent in the T cell-deficient athymic nude rats.ConclusionsThese data implicate MBP as a novel mediator of pain. Furthermore, the action of MMPs expressed within 1 day post-injury is critical to the generation of tactile allodynia, neuroinflammation, and the immunodominant MBP digest peptides in nerve. These MBP peptides initiate mechanical allodynia in both a T cell-dependent and -independent manner. In the course of Wallerian degeneration, the repeated exposure of the cryptic MBP epitopes, which are normally sheltered from immunosurveillance, may induce the MBP-specific T cell clones and a self-sustaining immune reaction, which may together contribute to the transition of acute pain into a chronic neuropathic pain state

    The MMP-9/TIMP-1 Axis Controls the Status of Differentiation and Function of Myelin-Forming Schwann Cells in Nerve Regeneration

    Get PDF
    Background: Myelinating Schwann cells (mSCs) form myelin in the peripheral nervous system. Because of the works by us and others, matrix metalloproteinase-9 (MMP-9) has recently emerged as an essential component of the Schwann cell signaling network during sciatic nerve regeneration. Methodology/Principal Findings: In the present study, using the genome-wide transcriptional profiling of normal and injured sciatic nerves in mice followed by extensive bioinformatics analyses of the data, we determined that an endogenous, specific MMP-9 inhibitor [tissue inhibitor of metalloproteinases (TIMP)-1] was a top up-regulated gene in the injured nerve. MMP-9 capture followed by gelatin zymography and Western blotting of the isolated samples revealed the presence of the MMP-9/TIMP-1 heterodimers and the activated MMP-9 enzyme in the injured nerve within the first 24 h post-injury. MMP-9 and TIMP-1 co-localized in mSCs. Knockout of the MMP-9 gene in mice resulted in elevated numbers of de-differentiated/immature mSCs in the damaged nerve. Our comparative studies using MMP-9 knockout and wild-type mice documented an aberrantly enhanced proliferative activity and, accordingly, an increased number of post-mitotic Schwann cells, short internodes and additional nodal abnormalities in remyelinated nerves of MMP-9 knockout mice. These data imply that during the first days post-injury MMP-9 exhibits a functionally important anti-mitogenic activity in the wildtype mice. Pharmacological inhibition of MMP activity suppressed the expression of Nav1.7/1.8 channels in the crushe

    Spinal activity of interleukin 6 mediates myelin basic protein-induced allodynia

    No full text
    Mechanosensory fibers are enveloped by myelin, a unique multilamellar membrane permitting saltatory neuronal conduction. Damage to myelin is thought to contribute to severe pain evoked by innocuous tactile stimulation (i.e., mechanical allodynia). Our earlier (Liu et al., 2012) and present data demonstrate that a single injection of a myelin basic protein-derived peptide (MBP84-104) into an intact sciatic nerve produces a robust and long-lasting (>30days) mechanical allodynia in female rats. The MBP84-104 peptide represents the immunodominant epitope and requires T cells to maintain allodynia. Surprisingly, only systemic gabapentin (a ligand of voltage-gated calcium channel Ξ±2Ξ΄1), but not ketorolac (COX inhibitor), lidocaine (sodium channel blocker) or MK801 (NMDA antagonist) reverse allodynia induced by the intrasciatic MBP84-104. The genome-wide transcriptional profiling of the sciatic nerve followed by the bioinformatics analyses of the expression changes identified interleukin (IL)-6 as the major cytokine induced by MBP84-104 in both the control and athymic T cell-deficient nude rats. The intrasciatic MBP84-104 injection resulted in both unilateral allodynia and unilateral IL-6 increase the segmental spinal cord (neurons and astrocytes). An intrathecal delivery of a function-blocking IL-6 antibody reduced the allodynia in part by the transcriptional effects in large-diameter primary afferents in DRG. Our data suggest that MBP regulates IL-6 expression in the nervous system and that the spinal IL-6 activity mediates nociceptive processing stimulated by the MBP epitopes released after damage or disease of the somatosensory nervous system

    MMP-9/TIMP-1 relationship in the sciatic nerve.

    No full text
    <p><b>A,</b> Left, Multiple murine MMP-9 species are detected by gelatin zymography. The total extracts (10 Β΅g total protein each) of the naΓ―ve (N), distal (D) and proximal (P) segments of the transected nerves were analyzed by gelatin zymography. Right lane, the purified MMP-9 sample isolated from an aliquot (25 Β΅g total protein) of the proximal nerve extract (P) is provided as a reference. Middle, the purified MMP-9 samples isolated from the naΓ―ve (N) and crushed (I) nerve (50 Β΅g and 25 Β΅g total protein, respectively) 1, 3, 6 and 24 h post-injury were analyzed by gelatin zymography. Note a 2Γ— protein loading of the naΓ―ve samples relative to the injured samples. Right, 2% agarose gel-electrophoresis of the RT-PCR products. The MMP-9 and TIMP-1 genes were amplified using the naive and crushed samples in the RT-PCR reactions. The unique primers were used in a 35-cycle PCR reaction to generate a 153 bp MMP-9 fragment, and 129 bp and 141 bp TIMP-1 fragments. GAPDH was used for sample normalization. Right lane (cDNA), the first strand cDNA control synthesized from the total mouse spleen RNA in the RT-PCR reaction. <b>B,</b> High molecular weight forms of MMP-9 in the injured sciatic nerve. The reduced (+DTT) and unreduced (βˆ’DTT) purified MMP-9 samples (15 Β΅g total protein each) isolated from the injured sciatic nerve 0, 1, 3, 6 and 24 h post-injury were analyzed by Western blotting with the MMP-9 antibody. <b>C,</b> Immunofluorescence for MMP-9 and TIMP-1 co-localize in mSCs of crushed nerve 24 h post-injury. Upper panel, MMP-9 (green) and TIMP-1 (red) co-localize in crescent structures of mSCs (arrows). Circular structures are MMP-9 but not TIMP-1 reactive (arrowheads). Lower panel, MMP-9 (red) localizes in the cytoplasm of mSCs, marked with MBP (green). MMP-9 is also detected in axoplasm of mSCs (arrows). Scale bar, 10 Β΅m.</p

    TIMP-1: MMP-9 ratio in the nerve samples.

    No full text
    <p>The normalized signal intensity of the MMP-9 and TIMP-1 genes was derived from the genome-wide transcriptional profiling data. Note the dramatic increase of the TIMP-1∢ MMP-9 signal intensity ratio on days 1 and 5 post-injury (D1 and D5, respectively) in the peripheral nerve sample. DRG, dorsal root ganglion, day 5 post-injury.</p

    MNCV in MMP-9<sup>βˆ’/βˆ’</sup> and MMP-9<sup>+/+</sup> mice.

    No full text
    <p>Motor nerve conduction velocity (MNCV) expressed as mean Β± SEM (nβ€Š=β€Š7/group) analyzed with a two-tailed, unpaired t-test or an unpaired t-test with Welch's correction when variances were unequal. NS, not significant.</p

    MMPi therapy blocks Na<sub>v</sub>1.7 and 1.8 channel induction in nerve.

    No full text
    <p><i>Taqman</i> qPCR for Na<sub>v</sub>1.8 and 1.7 in normal or crushed (day 5) rat sciatic nerve normalized to GAPDH. GM6001 or vehicle was administered i.p. immediately and then daily at days 1–5 after crush. The mean fold increase Β± SEM to normal nerves (*, p<0.05; **, p<0.01, ANOVA, Tukey's post-hoc test of nβ€Š=β€Š6/group). NS, not significant.</p
    • …
    corecore