Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Neuroinflammation ; 21(1): 134, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802868

ABSTRACT

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


Subject(s)
Macrophages , Nerve Regeneration , Peripheral Nerve Injuries , Receptors, CCR2 , Wallerian Degeneration , Animals , Macrophages/metabolism , Macrophages/pathology , Mice , Nerve Regeneration/physiology , Wallerian Degeneration/pathology , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Receptors, CCR2/deficiency , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/metabolism , Mice, Inbred C57BL , Mice, Knockout , Sciatic Neuropathy/pathology , Axons/pathology , Mice, Transgenic , Disease Models, Animal , Sciatic Nerve/injuries , Sciatic Nerve/pathology , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism
2.
Prog Neurobiol ; 228: 102488, 2023 09.
Article in English | MEDLINE | ID: mdl-37355220

ABSTRACT

The role of inflammation in nervous system injury and disease is attracting increased attention. Much of that research has focused on microglia in the central nervous system (CNS) and macrophages in the peripheral nervous system (PNS). Much less attention has been paid to the roles played by neutrophils. Neutrophils are part of the granulocyte subtype of myeloid cells. These cells, like macrophages, originate and differentiate in the bone marrow from which they enter the circulation. After tissue damage or infection, neutrophils are the first immune cells to infiltrate into tissues and are directed there by specific chemokines, which act on chemokine receptors on neutrophils. We have reviewed here the basic biology of these cells, including their differentiation, the types of granules they contain, the chemokines that act on them, the subpopulations of neutrophils that exist, and their functions. We also discuss tools available for identification and further study of neutrophils. We then turn to a review of what is known about the role of neutrophils in CNS and PNS diseases and injury, including stroke, Alzheimer's disease, multiple sclerosis, amyotrophic lateral sclerosis, spinal cord and traumatic brain injuries, CNS and PNS axon regeneration, and neuropathic pain. While in the past studies have focused on neutrophils deleterious effects, we will highlight new findings about their benefits. Studies on their actions should lead to identification of ways to modify neutrophil effects to improve health.


Subject(s)
Axons , Neutrophils , Humans , Axons/physiology , Nerve Regeneration/physiology , Peripheral Nervous System , Biology
3.
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
4.
eNeuro ; 9(1)2022.
Article in English | MEDLINE | ID: mdl-35131866

ABSTRACT

Regeneration can occur in peripheral neurons after injury, but the mechanisms involved are not fully delineated. Macrophages in dorsal root ganglia (DRGs) are involved in the enhanced regeneration that occurs after a conditioning lesion (CL), but how macrophages stimulate this response is not known. Oncomodulin (Ocm) has been proposed as a proregenerative molecule secreted by macrophages and neutrophils, is expressed in the DRG after axotomy, and stimulates neurite outgrowth by DRG neurons in culture. Wild-type (WT) and Ocm knock-out (KO) mice were used to investigate whether Ocm plays a role in the CL response in DRG neurons after sciatic nerve transection. Neurite outgrowth was measured after 24 and 48 h in explant culture 7 d after a CL. Sciatic nerve regeneration was also measured in vivo 7 d after a CL and 2 d after a subsequent sciatic nerve crush. The magnitude of the increased neurite outgrowth following a CL was significantly smaller in explants from Ocm KO mice than in explants from WT mice. In vivo after a CL, increased regeneration was found in WT animals but not in KO animals. Macrophage accumulation and levels of interleukin-6 (IL-6) mRNA were measured in axotomized DRG from WT and Ocm KO animals, and both were significantly higher than in sham-operated ganglia. At 6 h after axotomy, Il-6 mRNA was higher in WT than in Ocm KO mice. Our data support the hypothesis that Ocm plays a necessary role in producing a normal CL response and that its effects possibly result in part from stimulation of the expression of proregenerative macrophage cytokines such as IL-6.


Subject(s)
Ganglia, Spinal , Nerve Regeneration , Animals , Calcium-Binding Proteins , Ganglia, Spinal/metabolism , Mice , Mice, Knockout , Nerve Regeneration/physiology , Neurons/metabolism , Sciatic Nerve/metabolism
5.
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
6.
Prog Neurobiol ; 173: 102-121, 2019 02.
Article in English | MEDLINE | ID: mdl-30579784

ABSTRACT

Neuroinflammation has positive and negative effects. This review focuses on the roles of macrophage in the PNS. Transection of PNS axons leads to degeneration and clearance of the distal nerve and to changes in the region of the axotomized cell bodies. In both locations, resident and infiltrating macrophages are found. Macrophages enter these areas in response to expression of the chemokine CCL2 acting on the macrophage receptor CCR2. In the distal nerve, macrophages and other phagocytes are involved in clearance of axonal debris, which removes molecules that inhibit nerve regeneration. In the cell body region, macrophage trigger the conditioning lesion response, a process in which neurons increase their regeneration after a prior lesion. In mice in which the genes for CCL2 or CCR2 are deleted, neither macrophage infiltration nor the conditioning lesion response occurs in dorsal root ganglia (DRG). Macrophages exist in different phenotypes depending on their environment. These phenotypes have different effects on axonal clearance and neurite outgrowth. The mechanism by which macrophages affect neuronal cell bodies is still under study. Overexpression of CCL2 in DRG in uninjured animals leads to macrophage accumulation in the ganglia and to an increase in the growth potential of DRG neurons. This increased growth requires activation of neuronal STAT3. In contrast, in acute demyelinating neuropathies, macrophages are involved in stripping myelin from peripheral axons. The molecular mechanisms that trigger macrophage action after trauma and in autoimmune disease are receiving increased attention and should lead to avenues to promote regeneration and protect axonal integrity.


Subject(s)
Chemokines/metabolism , Macrophages/metabolism , Nerve Regeneration/physiology , Peripheral Nerve Injuries/metabolism , Animals , Humans , Macrophages/pathology , Peripheral Nerve Injuries/pathology
7.
J Neuroinflammation ; 15(1): 192, 2018 Jun 26.
Article in English | MEDLINE | ID: mdl-29945607

ABSTRACT

BACKGROUND: Neuroinflammation accompanies neural trauma and most neurological diseases. Axotomy in the peripheral nervous system (PNS) leads to dramatic changes in the injured neuron: the cell body expresses a distinct set of genes known as regeneration-associated genes, the distal axonal segment degenerates and its debris is cleared, and the axons in the proximal segment form growth cones and extend neurites. These processes are orchestrated in part by immune and other non-neuronal cells. Macrophages in ganglia play an integral role in supporting regeneration. Here, we explore further the molecular and cellular components of the injury-induced immune response within peripheral ganglia. METHODS: Adult male wild-type (WT) and Ccr2 -/- mice were subjected to a unilateral transection of the sciatic nerve and axotomy of the superior cervical ganglion (SCG). Antibody arrays were used to determine the expression of chemokines and cytokines in the dorsal root ganglion (DRG) and SCG. Flow cytometry and immunohistochemistry were utilized to identify the cellular composition of the injury-induced immune response within ganglia. RESULTS: Chemokine expression in the ganglia differed 48 h after nerve injury with a large increase in macrophage inflammatory protein-1γ in the SCG but not in the DRG, while C-C class chemokine ligand 2 was highly expressed in both ganglia. Differences between WT and Ccr2 -/- mice were also observed with increased C-C class chemokine ligand 6/C10 expression in the WT DRG compared to C-C class chemokine receptor 2 (CCR2)-/- DRG and increased CXCL5 expression in CCR2-/- SCG compared to WT. Diminished macrophage accumulation in the DRG and SCG of Ccr2 -/- mice was found compared to WT ganglia 7 days after nerve injury. Interestingly, neutrophils were found in the SCG but not in the DRG. Cytokine expression, measured 7 days after injury, differed between ganglion type and genotype. Macrophage activation was assayed by colabeling ganglia with the anti-inflammatory marker CD206 and the macrophage marker CD68, and an almost complete colocalization of the two markers was found in both ganglia. CONCLUSIONS: This study demonstrates both molecular and cellular differences in the nerve injury-induced immune response between DRG and SCG and between WT and Ccr2 -/- mice.


Subject(s)
Cytokines/metabolism , Ganglia, Spinal/pathology , Gene Expression Regulation/physiology , Sciatic Neuropathy/complications , Sciatic Neuropathy/pathology , Superior Cervical Ganglion/pathology , Animals , Axotomy/adverse effects , Disease Models, Animal , Flow Cytometry , Ganglia, Spinal/metabolism , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Superior Cervical Ganglion/metabolism , Time Factors
8.
J Neurosci ; 37(43): 10258-10277, 2017 10 25.
Article in English | MEDLINE | ID: mdl-28912156

ABSTRACT

Wallerian degeneration (WD) is considered an essential preparatory stage to the process of axonal regeneration. In the peripheral nervous system, infiltrating monocyte-derived macrophages, which use the chemokine receptor CCR2 to gain entry to injured tissues from the bloodstream, are purportedly necessary for efficient WD. However, our laboratory has previously reported that myelin clearance in the injured sciatic nerve proceeds unhindered in the Ccr2-/- mouse model. Here, we extensively characterize WD in male Ccr2-/- mice and identify a compensatory mechanism of WD that is facilitated primarily by neutrophils. In response to the loss of CCR2, injured Ccr2-/- sciatic nerves demonstrate prolonged expression of neutrophil chemokines, a concomitant extended increase in the accumulation of neutrophils in the nerve, and elevated phagocytosis by neutrophils. Neutrophil depletion substantially inhibits myelin clearance after nerve injury in both male WT and Ccr2-/- mice, highlighting a novel role for these cells in peripheral nerve degeneration that spans genotypes.SIGNIFICANCE STATEMENT The accepted view in the basic and clinical neurosciences is that the clearance of axonal and myelin debris after a nerve injury is directed primarily by inflammatory CCR2+ macrophages. However, we demonstrate that this clearance is nearly identical in WT and Ccr2-/- mice, and that neutrophils replace CCR2+ macrophages as the primary phagocytic cell. We find that neutrophils play a major role in myelin clearance not only in Ccr2-/- mice but also in WT mice, highlighting their necessity during nerve degeneration in the peripheral nervous system. These degeneration studies may propel improvements in nerve regeneration and draw critical parallels to mechanisms of nerve degeneration and regeneration in the CNS and in the context of peripheral neuropathies.


Subject(s)
Disease Models, Animal , Myelin Sheath/metabolism , Neutrophils/metabolism , Sciatic Neuropathy/metabolism , Wallerian Degeneration/metabolism , Animals , Female , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myelin Sheath/pathology , Nerve Crush/methods , Neutrophils/pathology , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/pathology , Phagocytosis/physiology , Random Allocation , Sciatic Neuropathy/pathology , Wallerian Degeneration/pathology
9.
Exp Neurol ; 296: 1-15, 2017 10.
Article in English | MEDLINE | ID: mdl-28645526

ABSTRACT

Neuropathy is a major diabetic complication. While the mechanism of this neuropathy is not well understood, it is believed to result in part from deficient nerve regeneration. Work from our laboratory established that gp130 family of cytokines are induced in animals after axonal injury and are involved in the induction of regeneration-associated genes (RAGs) and in the conditioning lesion response. Here, we examine whether a reduction of cytokine signaling occurs in diabetes. Streptozotocin (STZ) was used to destroy pancreatic ß cells, leading to chronic hyperglycemia. Mice were injected with either low doses of STZ (5×60mg/kg) or a single high dose (1×200mg/kg) and examined after three or one month, respectively. Both low and high dose STZ treatment resulted in sustained hyperglycemia and functional deficits associated with the presence of both sensory and autonomic neuropathy. Diabetic mice displayed significantly reduced intraepidermal nerve fiber density and sudomotor function. Furthermore, low and high dose diabetic mice showed significantly reduced tactile touch sensation measured with Von Frey monofilaments. To look at the regenerative and injury-induced responses in diabetic mice, neurons in both superior cervical ganglia (SCG) and the 4th and 5th lumbar dorsal root ganglia (DRG) were unilaterally axotomized. Both high and low dose diabetic mice displayed significantly less axonal regeneration in the sciatic nerve, when measured in vivo, 48h after crush injury. Significantly reduced induction of two gp130 cytokines, leukemia inhibitory factor and interleukin-6, occurred in diabetic animals in SCG 6h after injury compared to controls. Injury-induced expression of interleukin-6 was also found to be significantly reduced in the DRG at 6h after injury in low and high dose diabetic mice. These effects were accompanied by reduced phosphorylation of signal transducer and activator of transcription 3 (STAT3), a downstream effector of the gp130 signaling pathway. We also found decreased induction of several gp130-dependent RAGs, including galanin and vasoactive intestinal peptide. Together, these data suggest a novel mechanism for the decreased response of diabetic sympathetic and sensory neurons to injury.


Subject(s)
Cytokine Receptor gp130/metabolism , Diabetes Mellitus, Experimental/pathology , Gene Expression Regulation/physiology , Nerve Degeneration/etiology , Signal Transduction/physiology , Superior Cervical Ganglion/metabolism , Animals , Antibiotics, Antineoplastic/toxicity , Blood Glucose/drug effects , Body Weight/drug effects , Cytokine Receptor gp130/genetics , Cytokines/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/complications , Disease Models, Animal , Fasting/blood , Hyperalgesia/etiology , Hyperglycemia/etiology , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/pathology , Nerve Tissue Proteins/metabolism , Pain Measurement , Signal Transduction/drug effects , Streptozocin/toxicity , Superior Cervical Ganglion/drug effects , Sweating/drug effects
11.
Exp Neurol ; 275 Pt 1: 25-37, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26431741

ABSTRACT

Neuroinflammation plays a critical role in the regeneration of peripheral nerves following axotomy. An injury to the sciatic nerve leads to significant macrophage accumulation in the L5 DRG, an effect not seen when the dorsal root is injured. We recently demonstrated that this accumulation around axotomized cell bodies is necessary for a peripheral conditioning lesion response to occur. Here we asked whether overexpression of the monocyte chemokine CCL2 specifically in DRG neurons of uninjured mice is sufficient to cause macrophage accumulation and to enhance regeneration or whether other injury-derived signals are required. AAV5-EF1α-CCL2 was injected intrathecally, and this injection led to a time-dependent increase in CCL2 mRNA expression and macrophage accumulation in L5 DRG, with a maximal response at 3 weeks post-injection. These changes led to a conditioning-like increase in neurite outgrowth in DRG explant and dissociated cell cultures. This increase in regeneration was dependent upon CCL2 acting through its primary receptor CCR2. When CCL2 was overexpressed in CCR2-/- mice, macrophage accumulation and enhanced regeneration were not observed. To address the mechanism by which CCL2 overexpression enhances regeneration, we tested for elevated expression of regeneration-associated genes in these animals. Surprisingly, we found that CCL2 overexpression led to a selective increase in LIF mRNA and neuronal phosphorylated STAT3 (pSTAT3) in L5 DRGs, with no change in expression seen in other RAGs such as GAP-43. Blockade of STAT3 phosphorylation by each of two different inhibitors prevented the increase in neurite outgrowth. Thus, CCL2 overexpression is sufficient to induce macrophage accumulation in uninjured L5 DRGs and increase the regenerative capacity of DRG neurons via a STAT3-dependent mechanism.


Subject(s)
Chemokine CCL2/metabolism , Ganglia, Spinal/metabolism , Nerve Regeneration/physiology , Neurites/metabolism , Neurons/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cells, Cultured , Chemokine CCL2/genetics , Ganglia, Spinal/cytology , Macrophages/metabolism , Mice , Neurons/cytology , Peripheral Nerve Injuries/metabolism , Phosphorylation , Sciatic Nerve/injuries , Sciatic Nerve/metabolism
12.
Mol Cell Biol ; 35(24): 4238-52, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26416880

ABSTRACT

Neurite outgrowth is key to the formation of functional circuits during neuronal development. Neurotrophins, including nerve growth factor (NGF), increase neurite outgrowth in part by altering the function and expression of Ca(2+)-permeable cation channels. Here we report that transient receptor potential vanilloid 2 (TRPV2) is an intracellular Ca(2+)-permeable TRPV channel upregulated by NGF via the mitogen-activated protein kinase (MAPK) signaling pathway to augment neurite outgrowth. TRPV2 colocalized with Rab7, a late endosome protein, in addition to TrkA and activated extracellular signal-regulated kinase (ERK) in neurites, indicating that the channel is closely associated with signaling endosomes. In line with these results, we showed that TRPV2 acts as an ERK substrate and identified the motifs necessary for phosphorylation of TRPV2 by ERK. Furthermore, neurite length, TRPV2 expression, and TRPV2-mediated Ca(2+) signals were reduced by mutagenesis of these key ERK phosphorylation sites. Based on these findings, we identified a previously uncharacterized mechanism by which ERK controls TRPV2-mediated Ca(2+) signals in developing neurons and further establish TRPV2 as a critical intracellular ion channel in neuronal function.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Nerve Growth Factor/metabolism , Neurites/metabolism , Neurons/metabolism , TRPV Cation Channels/metabolism , Animals , Calcium/metabolism , Calcium Channels/metabolism , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/genetics , HEK293 Cells , Humans , MAP Kinase Signaling System , Neurogenesis/physiology , Neurons/cytology , PC12 Cells , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , RNA Interference , RNA, Small Interfering , Rats , Receptor, trkA/metabolism , TRPV Cation Channels/genetics , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
13.
J Neurosci ; 33(41): 16236-48, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-24107955

ABSTRACT

Macrophages have been implicated in peripheral nerve regeneration for some time, supposedly through their involvement in Wallerian degeneration, the process by which the distal nerve degenerates after axotomy and is cleared by phagocytosis. Thus, in several studies in which macrophage accumulation in the distal nerve was reduced and Wallerian degeneration inhibited, regeneration was delayed. However, this interpretation ignores the more recent findings that macrophages also accumulate around axotomized cell bodies. The function of macrophage action at this second site has not been clear. In two mutant strains of mice, the slow Wallerian degeneration (Wld(s)) mouse and the chemokine receptor CCR2 knock-out mouse, we report that macrophage accumulation after axotomy was abolished in both the dorsal root ganglion (DRG) and the distal sciatic nerve. To measure neurite outgrowth, DRG neurons were given a conditioning lesion, and outgrowth was measured in vitro 7 d later in the absence of the distal nerve segment. The increased growth normally seen after a conditioning lesion did not occur or was reduced in Wld(s) or CCR2(-/-) mice. In the superior cervical ganglion (SCG), particularly in Wld(s) mice, macrophage accumulation was reduced but not abolished after axotomy. In SCG neurons from Wld(s) mice, the conditioning lesion response was unchanged; however, in CCR2(-/-) mice in which the effect on macrophage accumulation was greater, SCG neurite outgrowth was significantly reduced. These results indicate that macrophages affect neurite outgrowth by acting at the level of peripheral ganglia in addition to any effects they might produce by facilitation of Wallerian degeneration.


Subject(s)
Ganglia, Spinal/physiology , Macrophages , Nerve Regeneration/physiology , Neurites/physiology , Sciatic Nerve/physiology , Animals , Axotomy , Female , Ganglia, Spinal/injuries , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Neurons/physiology , Real-Time Polymerase Chain Reaction , Sciatic Nerve/injuries
14.
J Neurosci ; 33(11): 4867-74, 2013 Mar 13.
Article in English | MEDLINE | ID: mdl-23486957

ABSTRACT

The cervical sympathetic trunks (CSTs) contain axons of preganglionic neurons that innervate the superior cervical ganglia (SCGs). Because regeneration of CST fibers can be extensive and can reestablish certain specific patterns of SCG connections, restoration of end organ function would be expected. This expectation was examined with respect to the pineal gland, an organ innervated by the two SCGs. The activity of pineal serotonin N-acetyltransferase (NAT) exhibits a large circadian rhythm that is dependent on the sympathetic input of the gland, with high activity at night. Thirty-six hours after the CSTs were crushed bilaterally, nocturnal NAT was decreased by 99%. Three months later, enzyme activity had recovered only to 15% of control values, a recovery dependent on regeneration of CST fibers. Nevertheless, a small day/night rhythm was present in lesioned animals. Neither the density of the adrenergic innervation of the gland nor the ability of an adrenergic agonist to stimulate NAT activity was reduced in rats with regenerated CSTs. In addition, stimulation of the regenerated CST at a variety of frequencies was at least as effective in increasing NAT activity as seen with control nerves. These data suggest that the failure of pineal function to recover is not attributable to a quantitative deficit in the extent of reinnervation or synaptic efficacy. Rather, we suggest that there is some loss of specificity in the synaptic connections made in the SCG during reinnervation, resulting in a loss of the central neuronal information necessary for directing a normal NAT rhythm and thus normal pineal function.


Subject(s)
Autonomic Nervous System Diseases/pathology , Autonomic Nervous System Diseases/physiopathology , Axons/pathology , Nerve Regeneration/physiology , Pineal Gland/physiopathology , Superior Cervical Ganglion/pathology , Animals , Arylalkylamine N-Acetyltransferase/metabolism , Axons/drug effects , Biophysics , Brocresine/pharmacology , Circadian Rhythm/physiology , Disease Models, Animal , Dose-Response Relationship, Drug , Electric Stimulation , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Enzymologic/physiology , Isoproterenol/pharmacology , Male , Nordefrin/pharmacology , Pineal Gland/metabolism , Rats , Rats, Sprague-Dawley , Superior Cervical Ganglion/drug effects , Sympathomimetics/pharmacology , Time Factors , Tyrosine 3-Monooxygenase
16.
Mol Cell Neurosci ; 46(3): 671-80, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21241805

ABSTRACT

Functional noradrenergic transmission requires the coordinate expression of enzymes involved in norepinephrine (NE) synthesis, as well as the norepinephrine transporter (NET) which removes NE from the synapse. Inflammatory cytokines acting through gp130 can suppress the noradrenergic phenotype in sympathetic neurons. This occurs in a subset of sympathetic neurons during development and also occurs in adult neurons after injury. For example, cytokines suppress noradrenergic function in sympathetic neurons after axotomy and during heart failure. The molecular basis for suppression of noradrenergic genes is not well understood, but previous studies implicated a reduction of Phox2a in cytokine suppression of dopamine beta hydroxylase. We used sympathetic neurons and neuroblastoma cells to investigate the role of Phox2a in cytokine suppression of NET transcription. Chromatin immunoprecipitation experiments revealed that Phox2a did not bind the NET promoter, and overexpression of Phox2a did not prevent cytokine suppression of NET transcription. Hand2 and Gata3 are transcription factors that induce noradrenergic genes during development and are present in mature sympathetic neurons. Both Hand2 and Gata3 were decreased by cytokines in sympathetic neurons and neuroblastoma cells. Overexpression of either Hand2 or Gata3 was sufficient to rescue NET transcription following suppression by cytokines. We examined expression of these genes following axotomy to determine if their expression was altered following nerve injury. NET and Hand2 mRNAs decreased significantly in sympathetic neurons 48 h after axotomy, but Gata3 mRNA was unchanged. These data suggest that cytokines can inhibit NET expression through downregulation of Hand2 or Gata3 in cultured sympathetic neurons, but axotomy in adult animals selectively suppresses Hand2 expression.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cytokines/metabolism , Neurons/metabolism , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Animals , Axotomy , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Ciliary Neurotrophic Factor/metabolism , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Neurons/cytology , Norepinephrine Plasma Membrane Transport Proteins/genetics , Rats , Rats, Sprague-Dawley
17.
Front Mol Neurosci ; 4: 62, 2011.
Article in English | MEDLINE | ID: mdl-22319466

ABSTRACT

Adult peripheral neurons, in contrast to adult central neurons, are capable of regeneration after axonal damage. Much attention has focused on the changes that accompany this regeneration in two places, the distal nerve segment (where phagocytosis of axonal debris, changes in the surface properties of Schwann cells, and induction of growth factors and cytokines occur) and the neuronal cell body (where dramatic changes in cell morphology and gene expression occur). The changes in the axotomized cell body are often referred to as the "cell body response." The focus of the current review is a family of cytokines, the glycoprotein 130 (gp130) cytokines, which produce their actions through a common gp130 signaling receptor and which function as injury signals for axotomized peripheral neurons, triggering changes in gene expression and in neurite outgrowth. These cytokines play important roles in the responses of sympathetic, sensory, and motor neurons to injury. The best studied of these cytokines in this context are leukemia inhibitory factor (LIF) and interleukin (IL)-6, but experiments with conditional gp130 knockout animals suggest that other members of this family, not yet determined, are also involved. The primary gp130 signaling pathway shown to be involved is the activation of Janus kinase (JAK) and the transcription factors Signal Transducers and Activators of Transcription (STAT), though other downstream pathways such as mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) may also play a role. gp130 signaling may involve paracrine, retrograde, and autocrine actions of these cytokines. Recent studies suggest that manipulation of this cytokine system can also stimulate regeneration by injured central neurons.

18.
Dev Neurobiol ; 69(6): 392-400, 2009 May.
Article in English | MEDLINE | ID: mdl-19280647

ABSTRACT

Adult peripheral neurons exhibit dramatic changes in gene expression after axonal injury, including changes in neuropeptide phenotype. For example, sympathetic neurons in the superior cervical ganglion (SCG) begin to express vasoactive intestinal peptide (VIP), galanin, pituitary adenylate cyclase activating polypeptide (PACAP), and cholecystokinin after axotomy. Before these changes, nonneuronal cells in the SCG begin to express leukemia inhibitory factor (LIF). When the effects of axotomy were compared in LIF-/- and wild-type mice, the increases in VIP and galanin expression were less in the former, though significant increases still occurred. LIF belongs to a family of cytokines with overlapping physiological effects and multimeric receptors containing the subunit gp130. Real-time PCR revealed large increases in the SCG after axotomy in mRNA for three members of this cytokine family, interleukin (IL)-6, IL-11, and LIF, with modest increases in oncostatin M, no changes in ciliary neurotrophic factor, and decreases in cardiotrophin-1. To explore the role of these cytokines, animals with selective elimination of the gp130 receptor in noradrenergic neurons were studied. No significant changes in mRNA levels for VIP, galanin, and PACAP were seen in axotomized ganglia from these mutant mice, while the increase in cholecystokinin was as large as that seen in wild-type mice. The data indicate that the inductions of VIP, galanin, and PACAP after axotomy are completely dependent on gp130 cytokines and that a second cytokine, in addition to LIF, is involved. The increase in cholecystokinin after axotomy, however, does not require the action of these cytokines.


Subject(s)
Cytokine Receptor gp130/metabolism , Gene Expression Regulation/genetics , Neurons/metabolism , Neuropeptides/metabolism , Superior Cervical Ganglion/cytology , Activating Transcription Factor 3/genetics , Activating Transcription Factor 3/metabolism , Analysis of Variance , Animals , Axotomy/methods , Cholecystokinin/genetics , Cholecystokinin/metabolism , Cytokine Receptor gp130/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptides/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/genetics , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , RNA, Messenger/metabolism , STAT3 Transcription Factor/metabolism , Time Factors , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , Vasoactive Intestinal Peptide/genetics , Vasoactive Intestinal Peptide/metabolism
19.
Neuroreport ; 18(16): 1729-33, 2007 Oct 29.
Article in English | MEDLINE | ID: mdl-17921877

ABSTRACT

Sensory neurons show enhanced neurite outgrowth in vivo and in vitro following a conditioning lesion. Previous studies have shown that these effects are dependent on two members of the gp130 family of cytokines, leukemia inhibitory factor and interleukin-6. Here, we asked whether galanin, a neuropeptide induced by these cytokines, plays a role in the conditioning lesion response. Following a conditioning lesion, neurite outgrowth in culture was reduced in sensory neurons from galanin -/- mice compared with those from wild type controls. In neurons from wild type mice, the length of the longest neurite was increased 2.4-fold after a conditioning lesion, compared with 1.8-fold in neurons from knockout animals. The results indicate that the induction of galanin plays an important role in triggering the conditioning lesion response.


Subject(s)
Galanin/metabolism , Ganglia, Spinal/metabolism , Neurites/metabolism , Neurons, Afferent/metabolism , Animals , Axotomy , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Enlargement/drug effects , Cell Size , Cells, Cultured , Galanin/genetics , Ganglia, Spinal/cytology , Ganglia, Spinal/immunology , Immunohistochemistry , Interleukin-6/immunology , Interleukin-6/metabolism , Leukemia Inhibitory Factor/immunology , Leukemia Inhibitory Factor/metabolism , Mice , Mice, Knockout , Neurites/immunology , Neurites/ultrastructure , Neurons, Afferent/cytology , Neurons, Afferent/immunology , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/physiopathology
20.
Brain Res ; 1159: 119-23, 2007 Jul 23.
Article in English | MEDLINE | ID: mdl-17583680

ABSTRACT

Activating transcription factor 3 (ATF3) has been proposed as a marker for injured neurons. Thus, while undetectable normally in sensory, motor, or sympathetic neurons, ATF3-like immunoreactivity (ATF3-IR) is readily detectable in such cells after axotomy. Here we examined ATF3-IR in the superior cervical ganglion (SCG) and the middle and inferior cervical ganglia (MICG) after transection of the predominantly preganglionic cervical sympathetic trunk (CST). The purpose of the study was to determine whether neurons in the SCG would exhibit ATF3-IR after decentralization and, if they did not, whether the induction of ATF3-IR was sensitive enough to identify the small numbers of neurons in the SCG and MICG that project their axons into the CST. Following transection of the CST, the majority of deafferented neurons in the SCG showed no ATF3-IR; however, a small group of neurons in both the SCG and MICG were labeled, and the location of the labeled neurons within these ganglia corresponded to that of neurons axotomized by this procedure. Furthermore, the ATF3-positive neurons in the MICG could be retrogradely labeled from the transected CST. In addition, a large number of smaller cells were labeled in the SCG, though not in the MICG, and some of these cells were double labeled with an antiserum to the glial protein S-100. These data indicate that, after transection of the CST, neuronal labeling in the SCG and MICG is restricted to axotomized neurons but that in addition there is extensive labeling of glial cells associated with anterograde degeneration within the SCG.


Subject(s)
Activating Transcription Factor 3/metabolism , Autonomic Fibers, Preganglionic/pathology , Ganglia, Sympathetic/pathology , Neurons/metabolism , Animals , Axotomy/methods , Male , Rats , Rats, Sprague-Dawley , S100 Proteins/metabolism , Satellite Cells, Perineuronal/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...