Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Elife ; 122023 05 31.
Article in English | MEDLINE | ID: mdl-37254842

ABSTRACT

Resident macrophages are distributed across all tissues and are highly heterogeneous due to adaptation to different tissue-specific environments. The resident macrophages of the sensory ganglia (sensory neuron-associated macrophages, sNAMs) are in close contact with the cell body of primary sensory neurons and might play physiological and pathophysiological roles. After peripheral nerve injury, there is an increase in the population of macrophages in the sensory ganglia, which have been implicated in different conditions, including neuropathic pain development. However, it is still under debate whether macrophage accumulation in the sensory ganglia after peripheral nerve injury is due to the local proliferation of resident macrophages or a result of blood monocyte infiltration. Here, we confirmed that the number of macrophages increased in the sensory ganglia after the spared nerve injury (SNI) model in mice. Using different approaches, we found that the increase in the number of macrophages in the sensory ganglia after SNI is a consequence of the proliferation of resident CX3CR1+ macrophages, which participate in the development of neuropathic pain, but not due to infiltration of peripheral blood monocytes. These proliferating macrophages are the source of pro-inflammatory cytokines such as TNF and IL-1b. In addition, we found that CX3CR1 signaling is involved in the sNAMs proliferation and neuropathic pain development after peripheral nerve injury. In summary, these results indicated that peripheral nerve injury leads to sNAMs proliferation in the sensory ganglia in a CX3CR1-dependent manner accounting for neuropathic pain development. In conclusion, sNAMs proliferation could be modulated to change pathophysiological conditions such as chronic neuropathic pain.


Subject(s)
Neuralgia , Peripheral Nerve Injuries , Mice , Animals , Peripheral Nerve Injuries/complications , Ganglia, Spinal , Macrophages , Ganglia, Sensory , Sensory Receptor Cells , Cell Proliferation , Hyperalgesia
2.
Methods Cell Biol ; 168: 249-276, 2022.
Article in English | MEDLINE | ID: mdl-35366986

ABSTRACT

Due to the large volume of surgeries and the subsequent incidence of postsurgical pain, it is vital that the underlying mechanisms of postsurgical pain are thoroughly understood. The intensity of acute postsurgical pain is typically dependent on the severity of tissue damage the surgery produces, and the development of chronic pain is more frequent in major surgeries than in minor ones. It is therefore important that postsurgical pain studies are conducted with the differences between major and minor surgeries in mind. To this end, the paw incision and skin muscle incision and retraction models are the focus of this chapter as they feature aspects observed in minor and major surgeries in humans, respectively. Several elements of these models translate to humans with some limitations, as they allow for the measurement of reflexive, spontaneous, and functional pain-like behavior. For these attributes, the SMIR and paw incision surgeries are widely used in postsurgical pain research. Here we layout detailed protocols to instruct experienced as well as inexperienced researchers studying postsurgical pain in rats and mice.


Subject(s)
Chronic Pain , Rodentia , Animals , Chronic Pain/complications , Mice , Pain, Postoperative/etiology , Rats , Rats, Sprague-Dawley , Skin
3.
Methods Cell Biol ; 168: 277-298, 2022.
Article in English | MEDLINE | ID: mdl-35366987

ABSTRACT

While cancer patients may have chemotherapeutics to thank for being cured of their malignancy, they are often left to suffer a disabling neuropathy induced by that same cancer treatment. This neuropathy, known as chemotherapy-induced peripheral neuropathy, or CIPN, is one of the most debilitating survivorship concerns for patients, with many citing hallmark symptoms of hyperalgesia, allodynia, and numbness, and subsequently reducing their dose or even ceasing treatment altogether. Investigations into this interplay between the antineoplastic activity of chemotherapeutic agents and the preservation of peripheral nerve health are therefore crucial for the development of CIPN treatment and prevention methods. Responding to need, current literature is inundated with varying preclinical models of CIPN. This chapter thus seeks to provide a detailed and reliable methodology for the induction and assessment of CIPN in mice, using a preclinical model that is both reproducible and translatable to several aspects of the clinical phenotype. Specifically, this chapter lays out a model for intermittent low-dose paclitaxel induction of CIPN in C57BL/6J mice, and a testing of this induction via von Frey filament mechanical hypersensitivity assays, a mechanical hyposensitivity (numbness) assay, and a cold-thermal allodynia assay (acetone test). These protocols can easily be adjusted to fit the needs of individual CIPN experiments, as stated throughout the chapter.


Subject(s)
Antineoplastic Agents , Peripheral Nervous System Diseases , Animals , Antineoplastic Agents/toxicity , Humans , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Hyperalgesia/genetics , Mice , Mice, Inbred C57BL , Paclitaxel/adverse effects , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/drug therapy
4.
Neurosci Lett ; 755: 135941, 2021 06 11.
Article in English | MEDLINE | ID: mdl-33961945

ABSTRACT

It has become increasingly clear that the innate immune system plays an essential role in the generation of many types of neuropathic pain including that which accompanies cancer treatment. In this article we review current findings of the role of the innate immune system in contributing to cancer treatment pain at the distal endings of peripheral nerve, in the nerve trunk, in the dorsal root ganglion and in the spinal dorsal horn.


Subject(s)
Antineoplastic Agents/adverse effects , Immunity, Innate/immunology , Neuralgia/chemically induced , Neuralgia/immunology , Animals , Ganglia, Spinal/drug effects , Ganglia, Spinal/immunology , Humans , Immunity, Innate/drug effects , Neuralgia/pathology , Spinal Cord Dorsal Horn/drug effects , Spinal Cord Dorsal Horn/immunology
5.
Biochem Pharmacol ; 188: 114520, 2021 06.
Article in English | MEDLINE | ID: mdl-33741328

ABSTRACT

Abnormal outgrowth of sensory nerves is one of the important contributors to pain associated with cancer and its treatments. Primary neuronal cultures derived from dorsal root ganglia (DRG) have been widely used to study pain-associated signal transduction and electrical activity of sensory nerves. However, there are only a few studies using primary DRG neuronal culture to investigate neurite outgrowth alterations due to underlying cancer-related factors and chemotherapeutic agents. In this study, primary DRG sensory neurons derived from mouse, non-human primate, and human were established in serum and growth factor-free conditions. A bovine serum albumin gradient centrifugation method improved the separation of sensory neurons from satellite cells. The purified DRG neurons were able to maintain their heterogeneous subpopulations, and displayed an increase in neurite growth when exposed to cancer-derived conditioned medium, while they showed a reduction in neurite length when treated with a neurotoxic chemotherapeutic agent. Additionally, a semi-automated quantification method was developed to measure neurite length in an accurate and time-efficient manner. Finally, these exogenous factors altered the gene expression patterns of murine primary sensory neurons, which are related to nerve growth, and neuro-inflammatory pain and nociceptor development. Together, the primary DRG neuronal culture in combination with a semi-automated quantification method can be a useful tool for further understanding the impact of exogenous factors on the growth of sensory nerve fibers and gene expression changes in sensory neurons.


Subject(s)
Cancer Pain/physiopathology , Neuronal Outgrowth/physiology , Sensory Receptor Cells/physiology , A549 Cells , Adult , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Cancer Pain/drug therapy , Cancer Pain/etiology , Carcinoma, Lewis Lung/complications , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/physiopathology , Cells, Cultured , Female , Humans , Macaca mulatta , Male , Mice , Mice, Inbred C57BL , Neuronal Outgrowth/drug effects , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , Sensory Receptor Cells/drug effects
6.
J Exp Med ; 217(10)2020 10 05.
Article in English | MEDLINE | ID: mdl-32697823

ABSTRACT

Th17 cell differentiation and pathogenicity depend on metabolic reprogramming inducing shifts toward glycolysis. Here, we show that the pyruvate kinase M2 (PKM2), a glycolytic enzyme required for cancer cell proliferation and tumor progression, is a key factor mediating Th17 cell differentiation and autoimmune inflammation. We found that PKM2 is highly expressed throughout the differentiation of Th17 cells in vitro and during experimental autoimmune encephalomyelitis (EAE) development. Strikingly, PKM2 is not required for the metabolic reprogramming and proliferative capacity of Th17 cells. However, T cell-specific PKM2 deletion impairs Th17 cell differentiation and ameliorates symptoms of EAE by decreasing Th17 cell-mediated inflammation and demyelination. Mechanistically, PKM2 translocates into the nucleus and interacts with STAT3, enhancing its activation and thereby increasing Th17 cell differentiation. Thus, PKM2 acts as a critical nonmetabolic regulator that fine-tunes Th17 cell differentiation and function in autoimmune-mediated inflammation.


Subject(s)
Autoimmunity/physiology , Inflammation/metabolism , Pyruvate Kinase/physiology , STAT3 Transcription Factor/metabolism , Th17 Cells/physiology , Animals , Cell Differentiation , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Flow Cytometry , Fluorescent Antibody Technique , Mice , Mice, Inbred C57BL , Pyruvate Kinase/metabolism , Real-Time Polymerase Chain Reaction , Th17 Cells/metabolism
7.
Pain ; 161(8): 1730-1743, 2020 08.
Article in English | MEDLINE | ID: mdl-32701834

ABSTRACT

The inflammatory/immune response at the site of peripheral nerve injury participates in the pathophysiology of neuropathic pain. Nevertheless, little is known about the local regulatory mechanisms underlying peripheral nerve injury that counteracts the development of pain. Here, we investigated the contribution of regulatory T (Treg) cells to the development of neuropathic pain by using a partial sciatic nerve ligation model in mice. We showed that Treg cells infiltrate and proliferate in the site of peripheral nerve injury. Local Treg cells suppressed the development of neuropathic pain mainly through the inhibition of the CD4 Th1 response. Treg cells also indirectly reduced neuronal damage and neuroinflammation at the level of the sensory ganglia. Finally, we identified IL-10 signaling as an intrinsic mechanism by which Treg cells counteract neuropathic pain development. These results revealed Treg cells as important inhibitory modulators of the immune response at the site of peripheral nerve injury that restrains the development of neuropathic pain. In conclusion, the boosting of Treg cell function/activity might be explored as a possible interventional approach to reduce neuropathic pain development after peripheral nerve damage.


Subject(s)
Neuralgia , Peripheral Nerve Injuries , T-Lymphocytes, Regulatory , Animals , Hyperalgesia , Mice , Mice, Inbred C57BL , Peripheral Nerve Injuries/complications , Sciatic Nerve , Th1 Cells
8.
Science ; 365(6450)2019 07 19.
Article in English | MEDLINE | ID: mdl-31320508

ABSTRACT

Inositol-requiring enzyme 1[α] (IRE1[α])-X-box binding protein spliced (XBP1) signaling maintains endoplasmic reticulum (ER) homeostasis while controlling immunometabolic processes. Yet, the physiological consequences of IRE1α-XBP1 activation in leukocytes remain unexplored. We found that induction of prostaglandin-endoperoxide synthase 2 (Ptgs2/Cox-2) and prostaglandin E synthase (Ptges/mPGES-1) was compromised in IRE1α-deficient myeloid cells undergoing ER stress or stimulated through pattern recognition receptors. Inducible biosynthesis of prostaglandins, including the pro-algesic mediator prostaglandin E2 (PGE2), was decreased in myeloid cells that lack IRE1α or XBP1 but not other ER stress sensors. Functional XBP1 transactivated the human PTGS2 and PTGES genes to enable optimal PGE2 production. Mice that lack IRE1α-XBP1 in leukocytes, or that were treated with IRE1α inhibitors, demonstrated reduced pain behaviors in PGE2-dependent models of pain. Thus, IRE1α-XBP1 is a mediator of prostaglandin biosynthesis and a potential target to control pain.


Subject(s)
Dinoprostone/biosynthesis , Endoribonucleases/metabolism , Leukocytes/metabolism , Pain, Postoperative/metabolism , Protein Serine-Threonine Kinases/metabolism , Visceral Pain/metabolism , X-Box Binding Protein 1/metabolism , Animals , Cells, Cultured , Cyclooxygenase 2/genetics , Endoribonucleases/genetics , Humans , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Pain, Postoperative/genetics , Promoter Regions, Genetic , Prostaglandin-E Synthases/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Unfolded Protein Response , Visceral Pain/genetics , X-Box Binding Protein 1/genetics
9.
J Leukoc Biol ; 106(3): 541-551, 2019 09.
Article in English | MEDLINE | ID: mdl-31150565

ABSTRACT

The development of neuropathic pain after peripheral nerve injury involves neuroimmune-glial interactions in the spinal cord. However, whether the development of neuropathic pain depends on the infiltration of peripheral immune cells, such as monocytes, into the spinal cord parenchyma after peripheral nerve damage remains unclear. Here, we used a combination of different techniques such as transgenic reporter mouse (Cx3cr1GFP/+ and Ccr2RFP/+ mice), bone marrow chimeric mice, and parabiosis to investigate this issue in spared nerve injury (SNI) model. Herein, we provided robust evidence that, although microglial cells are activated/proliferate at the dorsal horn of the spinal cord after SNI, peripheral hematopoietic cells (including monocytes) are not able to infiltrate into the spinal cord parenchyma. Furthermore, there was no evidence of CCR2 expression in intrinsic cells of the spinal cord. However, microglial cells activation/proliferation in the spinal cord and mechanical allodynia after SNI were reduced in Ccr2-deficient mice. These results suggest that blood-circulating leukocytes cells are not able to infiltrate the spinal cord parenchyma after distal peripheral nerve injury. Nevertheless, they indicate that CCR2-expressing cells might be indirectly regulating microglia activation/proliferation in the spinal cord after SNI. In conclusion, our study supports that CCR2 inhibition could be explored as an interventional approach to reduce microglia activation and consequently neuropathic pain development after peripheral nerve injury.


Subject(s)
Leukocytes/pathology , Peripheral Nerve Injuries/blood , Peripheral Nerve Injuries/pathology , Spinal Cord/pathology , Animals , Cell Proliferation , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelium, Vascular/pathology , Female , Hematopoietic Stem Cells/metabolism , Hyperalgesia/blood , Hyperalgesia/complications , Hyperalgesia/immunology , Hyperalgesia/pathology , Male , Mice, Inbred C57BL , Microglia/pathology , Monocytes/pathology , Neuralgia/blood , Neuralgia/complications , Neuralgia/immunology , Neuralgia/pathology , Receptors, CCR2/deficiency , Receptors, CCR2/metabolism
10.
Front Immunol ; 10: 3059, 2019.
Article in English | MEDLINE | ID: mdl-32047492

ABSTRACT

Neuroimmune-glia interactions have been implicated in the development of neuropathic pain. Interleukin-27 (IL-27) is a cytokine that presents regulatory activity in inflammatory conditions of the central nervous system. Thus, we hypothesized that IL-27 would participate in the neuropathic pain process. Here, we found that neuropathic pain caused by peripheral nerve injury (spared nerve injury model; SNI), was enhanced in IL-27-deficient(-/-) mice, whereas nociceptive pain is similar to that of wild-type mice. SNI induced an increase in the expression of IL-27 and its receptor subunit (Wsx1) in the sensory ganglia and spinal cord. IL-27 receptor was expressed mainly in resident macrophage, microglia, and astrocytes of the sensory ganglia and spinal cord, respectively. Finally, we identify that the antinociceptive effect of IL-27 was not observed in IL-10-/- mice. These results provided evidence that IL-27 is a cytokine produced after peripheral nerve injury that counteracts neuropathic pain development through induction of the antinociceptive cytokine IL-10. In summary, our study unraveled the role of IL-27 as a regulatory cytokine that counteracts the development of neuropathic pain after peripheral nerve damage. In conclusion, they indicate that immunotherapies based on IL-27 could emerge as possible therapeutic approaches for the prevention of neuropathic pain development after peripheral nerve injury.


Subject(s)
Disease Susceptibility , Interleukin-10/metabolism , Interleukin-27/metabolism , Neuralgia/etiology , Neuralgia/metabolism , Animals , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Ganglia, Spinal , Interleukin-27/genetics , Male , Mice , Mice, Knockout , Microglia/metabolism , Peripheral Nerve Injuries/complications , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Spinal Cord/metabolism , Spinal Cord/physiopathology
11.
Pain ; 160(1): 102-116, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30169421

ABSTRACT

Neuropathic pain is one of the most important types of chronic pain. It is caused by neuronal damage. Clinical and experimental studies suggest a critical role for neuroimmune interactions in the development of neuropathic pain. In this article, we have shown that the cytoplasmic receptor Nod-like receptor-2, NOD2, and its adaptor-signaling molecule RIPK2 participate in the development of neuropathic pain after peripheral nerve injury (spared nerve injury model). The activation of NOD2 signaling in peripheral macrophage mediates the development of neuropathic pain through the production of pronociceptive cytokines (tumor necrosis factor and IL-1ß). This study found that peripheral nerve injury promoted a systemic increase in the NOD2 ligand. These results highlight a previously undetermined role for NOD2 signaling in the development of neuropathic pain, suggesting a new potential target for preventing neuropathic pain.


Subject(s)
Macrophages/metabolism , Neuralgia/pathology , Neuralgia/physiopathology , Nod2 Signaling Adaptor Protein/metabolism , Animals , Bone Marrow Transplantation , Carrageenan/toxicity , Disease Models, Animal , Inflammation/chemically induced , Inflammation/therapy , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Minocycline/therapeutic use , Neuralgia/genetics , Neuralgia/surgery , Neuroprotective Agents/therapeutic use , Nod2 Signaling Adaptor Protein/genetics , RNA, Small Interfering/therapeutic use , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Xanthines/therapeutic use
12.
Sci Rep ; 5: 14648, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26511791

ABSTRACT

The assessment of articular nociception in experimental animals is a challenge because available methods are limited and subject to investigator influence. In an attempt to solve this problem, the purpose of this study was to establish the use of dynamic weight bearing (DWB) as a new device for evaluating joint nociception in an experimental model of antigen-induced arthritis (AIA) in mice. AIA was induced in Balb/c and C57BL/6 mice, and joint nociception was evaluated by DWB. Western Blotting and real-time PCR were used to determine protein and mRNA expression, respectively. DWB detected a dose- and time-dependent increase in joint nociception during AIA and was able to detect the dose-response effects of different classes of analgesics. Using DWB, it was possible to evaluate the participation of spinal glial cells (microglia and astrocytes) and cytokines (IL-1ß and TNFα) for the genesis of joint nociception during AIA. In conclusion, the present results indicated that DWB is an effective, objective and predictable test to study both the pathophysiological mechanisms involved in arthritic nociception in mice and for evaluating novel analgesic drugs against arthritis.


Subject(s)
Arthritis, Experimental/immunology , Arthritis, Experimental/physiopathology , Astrocytes/immunology , Microglia/immunology , Nociception , Analgesics/pharmacology , Animals , Arthritis, Experimental/pathology , Astrocytes/pathology , Interleukin-1beta/immunology , Male , Mice , Mice, Inbred BALB C , Microglia/pathology , Tumor Necrosis Factor-alpha/immunology , Weight-Bearing
SELECTION OF CITATIONS
SEARCH DETAIL
...