Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Neurosci ; 38(32): 7032-7057, 2018 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-29976627

RESUMEN

Injury, inflammation, and nerve damage initiate a wide variety of cellular and molecular processes that culminate in hyperexcitation of sensory nerves, which underlies chronic inflammatory and neuropathic pain. Using behavioral readouts of pain hypersensitivity induced by angiotensin II (Ang II) injection into mouse hindpaws, our study shows that activation of the type 2 Ang II receptor (AT2R) and the cell-damage-sensing ion channel TRPA1 are required for peripheral mechanical pain sensitization induced by Ang II in male and female mice. However, we show that AT2R is not expressed in mouse and human dorsal root ganglia (DRG) sensory neurons. Instead, expression/activation of AT2R on peripheral/skin macrophages (MΦs) constitutes a critical trigger of mouse and human DRG sensory neuron excitation. Ang II-induced peripheral mechanical pain hypersensitivity can be attenuated by chemogenetic depletion of peripheral MΦs. Furthermore, AT2R activation in MΦs triggers production of reactive oxygen/nitrogen species, which trans-activate TRPA1 on mouse and human DRG sensory neurons via cysteine modification of the channel. Our study thus identifies a translatable immune cell-to-sensory neuron signaling crosstalk underlying peripheral nociceptor sensitization. This form of cell-to-cell signaling represents a critical peripheral mechanism for chronic pain and thus identifies multiple druggable analgesic targets.SIGNIFICANCE STATEMENT Pain is a widespread health problem that is undermanaged by currently available analgesics. Findings from a recent clinical trial on a type II angiotensin II receptor (AT2R) antagonist showed effective analgesia for neuropathic pain. AT2R antagonists have been shown to reduce neuropathy-, inflammation- and bone cancer-associated pain in rodents. We report that activation of AT2R in macrophages (MΦs) that infiltrate the site of injury, but not in sensory neurons, triggers an intercellular redox communication with sensory neurons via activation of the cell damage/pain-sensing ion channel TRPA1. This MΦ-to-sensory neuron crosstalk results in peripheral pain sensitization. Our findings provide an evidence-based mechanism underlying the analgesic action of AT2R antagonists, which could accelerate the development of efficacious non-opioid analgesic drugs for multiple pain conditions.


Asunto(s)
Angiotensina II/fisiología , Hiperalgesia/fisiopatología , Macrófagos Peritoneales/metabolismo , Neuralgia/fisiopatología , Receptor de Angiotensina Tipo 2/fisiología , Células Receptoras Sensoriales/fisiología , Canal Catiónico TRPA1/fisiología , Angiotensina II/toxicidad , Antagonistas de Receptores de Angiotensina/farmacología , Animales , Comunicación Celular/fisiología , Células Cultivadas , Femenino , Ganglios Espinales/citología , Genes Reporteros , Humanos , Hiperalgesia/inducido químicamente , Hiperalgesia/tratamiento farmacológico , Imidazoles/farmacología , Activación de Macrófagos , Macrófagos Peritoneales/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuralgia/tratamiento farmacológico , Activación Neutrófila , Oxidación-Reducción , Piridinas/farmacología , Receptor de Angiotensina Tipo 2/genética , Células Receptoras Sensoriales/química , Piel/citología , Canal Catiónico TRPA1/deficiencia , Tacrolimus/análogos & derivados , Tacrolimus/farmacología
2.
Mol Pain ; 11: 1, 2015 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-25558916

RESUMEN

BACKGROUND: Itch-producing compounds stimulate receptors expressed on small diameter fibers that innervate the skin. Many of the currently known pruritogen receptors are Gq Protein-Coupled Receptors (GqPCR), which activate Protein Kinase C (PKC). Specific isoforms of PKC have been previously shown to perform selective functions; however, the roles of PKC isoforms in regulating itch remain unclear. In this study, we investigated the novel PKC isoform PKCδ as an intracellular modulator of itch signaling in response to histamine and the non-histaminergic pruritogens chloroquine and ß-alanine. RESULTS: Behavioral experiments indicate that PKCδ knock-out (KO) mice have a 40% reduction in histamine-induced scratching when compared to their wild type littermates. On the other hand, there were no differences between the two groups in scratching induced by the MRGPR agonists chloroquine or ß-alanine. PKCδ was present in small diameter dorsal root ganglion (DRG) neurons. Of PKCδ-expressing neurons, 55% also stained for the non-peptidergic marker IB4, while a smaller percentage (15%) expressed the peptidergic marker CGRP. Twenty-nine percent of PKCδ-expressing neurons also expressed TRPV1. Calcium imaging studies of acutely dissociated DRG neurons from PKCδ-KO mice show a 40% reduction in the total number of neurons responsive to histamine. In contrast, there was no difference in the number of capsaicin-responsive neurons between KO and WT animals. Acute pharmacological inhibition of PKCδ with an isoform-specific peptide inhibitor (δV1-1) also significantly reduced the number of histamine-responsive sensory neurons. CONCLUSIONS: Our findings indicate that PKCδ plays a role in mediating histamine-induced itch, but may be dispensable for chloroquine- and ß-alanine-induced itch.


Asunto(s)
Regulación hacia Abajo/genética , Histamina/efectos adversos , Proteína Quinasa C-alfa/metabolismo , Prurito/inducido químicamente , Prurito/metabolismo , Animales , Péptido Relacionado con Gen de Calcitonina , Calcio/metabolismo , Capsaicina/farmacología , Células Cultivadas , Cloroquina/efectos adversos , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Ganglios Espinales/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Quinasa C-alfa/genética , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , beta-Alanina/efectos adversos
3.
Mult Scler Relat Disord ; 45: 102439, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32769063

RESUMEN

BACKGROUND: Development of long-term immunologic memory relies upon humoral and cellular immune responses. Vaccinations aim to stimulate these responses against pathogens. Several studies have evaluated the impact of multiple sclerosis disease-modifying therapies on immune response to vaccines. Findings from these studies have important implications for people with multiple sclerosis who require vaccination and are using disease-modifying therapies. METHODS: Searches using PubMed and other engines were conducted in May 2020 to collect studies evaluating the impact of various disease-modifying therapies on immune responses to vaccination. RESULTS: Several studies demonstrated preserved immune responses in people treated with beta-interferons to multiple vaccine types. Limited data suggest vaccine responses to be preserved with dimethyl fumarate treatment, as well. Vaccine responses were reduced to varying degrees in those treated with glatiramer acetate, teriflunomide, sphingosine-1-phosphate receptor modulators, and natalizumab. The timing of vaccination played an important role in those treated with alemtuzumab. Humoral vaccine responses were significantly impaired by B cell depleting anti-CD20 monoclonal antibody therapies, particularly to a neoantigen. Data are lacking on vaccine responses in patients with multiple sclerosis taking cladribine and high-dose corticosteroids. Notably, the majority of these studies have focused on humoral responses, with few examining cellular immune responses to vaccination. CONCLUSIONS: Prior investigations into the effects of individual disease-modifying therapies on immune responses to existing vaccines can serve as a guide to expected responses to a SARS-CoV-2 vaccine. Responses to any vaccination depend on the vaccine type, the type of response (recall versus response to a novel antigen), and the impact of the individual disease-modifying therapy on humoral and cellular immunity in response to that vaccine type. When considering a given therapy, clinicians should weigh its efficacy against MS for the individual patient versus potential impact on responses to vaccinations that may be needed in the future.


Asunto(s)
Infecciones por Coronavirus/prevención & control , Huésped Inmunocomprometido/efectos de los fármacos , Huésped Inmunocomprometido/inmunología , Esclerosis Múltiple/inmunología , Pandemias/prevención & control , Neumonía Viral/prevención & control , Vacunas Virales/inmunología , Betacoronavirus/inmunología , COVID-19 , Vacunas contra la COVID-19 , Infecciones por Coronavirus/inmunología , Humanos , Inmunosupresores/uso terapéutico , Esclerosis Múltiple/tratamiento farmacológico , Neumonía Viral/inmunología , SARS-CoV-2
4.
eNeuro ; 5(2)2018.
Artículo en Inglés | MEDLINE | ID: mdl-29662945

RESUMEN

The use of human tissue to validate putative analgesic targets identified in rodents is a promising strategy for improving the historically poor translational record of preclinical pain research. We recently demonstrated that in mouse and human sensory neurons, agonists for metabotropic glutamate receptors 2 and 3 (mGluR2/3) reduce membrane hyperexcitability produced by the inflammatory mediator prostaglandin E2 (PGE2). Previous rodent studies indicate that mGluR2/3 can also reduce peripheral sensitization by suppressing inflammation-induced sensitization of TRPV1. Whether this observation similarly translates to human sensory neurons has not yet been tested. We found that activation of mGluR2/3 with the agonist APDC suppressed PGE2-induced sensitization of TRPV1 in mouse, but not human, sensory neurons. We also evaluated sensory neuron expression of the gene transcripts for mGluR2 (Grm2), mGluR3 (Grm3), and TRPV1 (Trpv1). The majority of Trpv1+ mouse and human sensory neurons expressed Grm2 and/or Grm3, and in both mice and humans, Grm2 was expressed in a greater percentage of sensory neurons than Grm3. Although we demonstrated a functional difference in the modulation of TRPV1 sensitization by mGluR2/3 activation between mouse and human, there were no species differences in the gene transcript colocalization of mGluR2 or mGluR3 with TRPV1 that might explain this functional difference. Taken together with our previous work, these results suggest that mGluR2/3 activation suppresses only some aspects of human sensory neuron sensitization caused by PGE2. These differences have implications for potential healthy human voluntary studies or clinical trials evaluating the analgesic efficacy of mGluR2/3 agonists or positive allosteric modulators.


Asunto(s)
Ganglios Espinales/metabolismo , Percepción del Dolor , Dolor/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Células Receptoras Sensoriales/metabolismo , Canales Catiónicos TRPV/metabolismo , Adolescente , Adulto , Animales , Técnicas de Cultivo de Célula , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Adulto Joven
5.
Nat Protoc ; 11(10): 1877-88, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27606776

RESUMEN

Primary cultures of rodent sensory neurons are widely used to investigate the cellular and molecular mechanisms involved in pain, itch, nerve injury and regeneration. However, translation of these preclinical findings may be greatly improved by direct validation in human tissues. We have developed an approach to extract and culture human sensory neurons in collaboration with a local organ procurement organization (OPO). Here we describe the surgical procedure for extraction of human dorsal root ganglia (hDRG) and the necessary modifications to existing culture techniques to prepare viable adult human sensory neurons for functional studies. Dissociated sensory neurons can be maintained in culture for >10 d, and they are amenable to electrophysiological recording, calcium imaging and viral gene transfer. The entire process of extraction and culturing can be completed in <7 h, and it can be performed by trained graduate students. This approach can be applied at any institution with access to organ donors consenting to tissue donation for research, and is an invaluable resource for improving translational research.


Asunto(s)
Ganglios Espinales/citología , Ganglios Espinales/cirugía , Células Receptoras Sensoriales/citología , Obtención de Tejidos y Órganos/métodos , Calcio/metabolismo , Técnicas de Cultivo de Célula/métodos , Separación Celular/métodos , Células Cultivadas , Técnicas de Transferencia de Gen , Humanos , Células Receptoras Sensoriales/metabolismo , Transducción Genética/métodos , Virus/genética
6.
Pain ; 157(9): 2081-2088, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27218869

RESUMEN

We introduce a strategy for preclinical research wherein promising targets for analgesia are tested in rodent and subsequently validated in human sensory neurons. We evaluate group II metabotropic glutamate receptors, the activation of which is efficacious in rodent models of pain. Immunohistochemical analysis showed positive immunoreactivity for mGlu2 in rodent dorsal root ganglia (DRG), peripheral fibers in skin, and central labeling in the spinal dorsal horn. We also found mGlu2-positive immunoreactivity in human neonatal and adult DRG. RNA-seq analysis of mouse and human DRG revealed a comparative expression profile between species for group II mGluRs and for opioid receptors. In rodent sensory neurons under basal conditions, activation of group II mGluRs with a selective group II agonist produced no changes to membrane excitability. However, membrane hyperexcitability in sensory neurons exposed to the inflammatory mediator prostaglandin E2 (PGE2) was prevented by (2R,4R)-4-aminopyrrolidine-2,4-dicarboxylate (APDC). In human sensory neurons from donors without a history of chronic pain, we show that PGE2 produced hyperexcitability that was similarly blocked by group II mGluR activation. These results reveal a mechanism for peripheral analgesia likely shared by mice and humans and demonstrate a translational research strategy to improve preclinical validation of novel analgesics using cultured human sensory neurons.


Asunto(s)
Neuronas/metabolismo , Nociceptores/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/genética , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Células Cultivadas , Dinoprostona/farmacología , Agonistas de Aminoácidos Excitadores , Ganglios Espinales/citología , Humanos , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/metabolismo , Tiempo de Reacción/efectos de los fármacos , Tiempo de Reacción/genética , Receptores de Glutamato Metabotrópico/genética , Tubulina (Proteína)/metabolismo
7.
J Pain ; 16(4): 346-56, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25640289

RESUMEN

UNLABELLED: Chronic pruritic conditions are often associated with dry skin and loss of epidermal barrier integrity. In this study, repeated application of acetone and ether followed by water (AEW) to the cheek skin of mice produced persistent scratching behavior with no increase in pain-related forelimb wiping, indicating the generation of itch without pain. Cheek skin immunohistochemistry showed a 64.5% increase in total epidermal innervation in AEW-treated mice compared to water-treated controls. This increase was independent of scratching, because mice prevented from scratching by Elizabethan collars showed similar hyperinnervation. To determine the effects of dry skin treatment on specific subsets of peripheral fibers, we examined Ret-positive, calcitonin gene-related peptide (CGRP)-positive, and glial cell line-derived neurotrophic factor family receptor α3 (GFRα3)-positive intraepidermal fiber density. AEW treatment increased Ret-positive fibers but not CGRP-positive or GFRα3-positive fibers, suggesting that a specific subset of nonpeptidergic fibers could contribute to dry skin itch. To test whether trigeminal ganglion neurons innervating the cheek exhibited altered excitability after AEW treatment, primary cultures of retrogradely labeled neurons were examined using whole-cell patch clamp electrophysiology. AEW treatment produced no differences in measures of excitability compared to water-treated controls. In contrast, a significantly higher proportion of trigeminal ganglion neurons was responsive to the nonhistaminergic pruritogen chloroquine after AEW treatment. We conclude that nonpeptidergic, Ret-positive fibers and chloroquine-sensitive neurons may contribute to dry skin pruritus. PERSPECTIVE: This study examines the underlying neurobiological mechanisms of persistent dry skin itch. Our results indicate that nonpeptidergic epidermal hyperinnervation and nonhistaminergic pruritic receptors are potential targets for chronic pruritus.


Asunto(s)
Cloroquina/toxicidad , Neuronas/fisiología , Prurito/fisiopatología , Piel/inervación , Ganglio del Trigémino/fisiopatología , Animales , Péptido Relacionado con Gen de Calcitonina/metabolismo , Calcio/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Cara/inervación , Cara/fisiopatología , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Técnicas de Trazados de Vías Neuroanatómicas , Neuronas/efectos de los fármacos , Neuronas/patología , Técnicas de Placa-Clamp , Proteínas Proto-Oncogénicas c-ret/genética , Proteínas Proto-Oncogénicas c-ret/metabolismo , Prurito/inducido químicamente , Prurito/etiología , Prurito/patología , Piel/efectos de los fármacos , Piel/fisiopatología , Ganglio del Trigémino/efectos de los fármacos , Ganglio del Trigémino/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA