Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 164
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(7): 1589-1616, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38552609

RESUMEN

The last 50 years have witnessed extraordinary developments in understanding mechanisms of carcinogenesis, synthesized as the hallmarks of cancer. Despite this logical framework, our understanding of the molecular basis of systemic manifestations and the underlying causes of cancer-related death remains incomplete. Looking forward, elucidating how tumors interact with distant organs and how multifaceted environmental and physiological parameters impinge on tumors and their hosts will be crucial for advances in preventing and more effectively treating human cancers. In this perspective, we discuss complexities of cancer as a systemic disease, including tumor initiation and promotion, tumor micro- and immune macro-environments, aging, metabolism and obesity, cancer cachexia, circadian rhythms, nervous system interactions, tumor-related thrombosis, and the microbiome. Model systems incorporating human genetic variation will be essential to decipher the mechanistic basis of these phenomena and unravel gene-environment interactions, providing a modern synthesis of molecular oncology that is primed to prevent cancers and improve patient quality of life and cancer outcomes.


Asunto(s)
Neoplasias , Humanos , Carcinogénesis , Microbiota , Neoplasias/genética , Neoplasias/patología , Neoplasias/terapia , Obesidad/complicaciones , Calidad de Vida
2.
Cell ; 165(7): 1749-1761, 2016 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-27315482

RESUMEN

Neurons are well suited for computations on millisecond timescales, but some neuronal circuits set behavioral states over long time periods, such as those involved in energy homeostasis. We found that multiple types of hypothalamic neurons, including those that oppositely regulate body weight, are specialized as near-perfect synaptic integrators that summate inputs over extended timescales. Excitatory postsynaptic potentials (EPSPs) are greatly prolonged, outlasting the neuronal membrane time-constant up to 10-fold. This is due to the voltage-gated sodium channel Nav1.7 (Scn9a), previously associated with pain-sensation but not synaptic integration. Scn9a deletion in AGRP, POMC, or paraventricular hypothalamic neurons reduced EPSP duration, synaptic integration, and altered body weight in mice. In vivo whole-cell recordings in the hypothalamus confirmed near-perfect synaptic integration. These experiments show that integration of synaptic inputs over time by Nav1.7 is critical for body weight regulation and reveal a mechanism for synaptic control of circuits regulating long term homeostatic functions.


Asunto(s)
Mantenimiento del Peso Corporal , Hipotálamo/citología , Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Neuronas/metabolismo , Sinapsis , Proteína Relacionada con Agouti/metabolismo , Animales , Homeostasis , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Transgénicos
3.
Proc Natl Acad Sci U S A ; 120(17): e2211631120, 2023 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-37071676

RESUMEN

Fibromyalgia is a debilitating widespread chronic pain syndrome that occurs in 2 to 4% of the population. The prevailing view that fibromyalgia results from central nervous system dysfunction has recently been challenged with data showing changes in peripheral nervous system activity. Using a mouse model of chronic widespread pain through hyperalgesic priming of muscle, we show that neutrophils invade sensory ganglia and confer mechanical hypersensitivity on recipient mice, while adoptive transfer of immunoglobulin, serum, lymphocytes, or monocytes has no effect on pain behavior. Neutrophil depletion abolishes the establishment of chronic widespread pain in mice. Neutrophils from patients with fibromyalgia also confer pain on mice. A link between neutrophil-derived mediators and peripheral nerve sensitization is already established. Our observations suggest approaches for targeting fibromyalgia pain via mechanisms that cause altered neutrophil activity and interactions with sensory neurons.


Asunto(s)
Dolor Crónico , Fibromialgia , Humanos , Neutrófilos , Hiperalgesia , Ganglios Sensoriales
4.
Brain ; 146(9): 3851-3865, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37222214

RESUMEN

Chronic pain affects millions of people worldwide and new treatments are needed urgently. One way to identify novel analgesic strategies is to understand the biological dysfunctions that lead to human inherited pain insensitivity disorders. Here we report how the recently discovered brain and dorsal root ganglia-expressed FAAH-OUT long non-coding RNA (lncRNA) gene, which was found from studying a pain-insensitive patient with reduced anxiety and fast wound healing, regulates the adjacent key endocannabinoid system gene FAAH, which encodes the anandamide-degrading fatty acid amide hydrolase enzyme. We demonstrate that the disruption in FAAH-OUT lncRNA transcription leads to DNMT1-dependent DNA methylation within the FAAH promoter. In addition, FAAH-OUT contains a conserved regulatory element, FAAH-AMP, that acts as an enhancer for FAAH expression. Furthermore, using transcriptomic analyses in patient-derived cells we have uncovered a network of genes that are dysregulated from disruption of the FAAH-FAAH-OUT axis, thus providing a coherent mechanistic basis to understand the human phenotype observed. Given that FAAH is a potential target for the treatment of pain, anxiety, depression and other neurological disorders, this new understanding of the regulatory role of the FAAH-OUT gene provides a platform for the development of future gene and small molecule therapies.


Asunto(s)
ARN Largo no Codificante , Humanos , Dolor/genética , Analgésicos , Ganglios Espinales
5.
Brain ; 146(10): 4033-4039, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37249190

RESUMEN

Melzak and Wall's gate control theory proposed that innocuous input into the dorsal horn of the spinal cord represses pain-inducing nociceptive input. Here we show that input from proprioceptive parvalbumin-expressing sensory neurons tonically represses nociceptor activation within dorsal root ganglia. Deletion of parvalbumin-positive sensory neurons leads to enhanced nociceptor activity measured with GCaMP3, increased input into wide dynamic range neurons of the spinal cord and increased acute and spontaneous pain behaviour, as well as potentiated innocuous sensation. Parvalbumin-positive sensory neurons express the enzymes and transporters necessary to produce vesicular GABA that is known to be released from depolarized somata. These observations support the view that gate control mechanisms occur peripherally within dorsal root ganglia.


Asunto(s)
Parvalbúminas , Células Receptoras Sensoriales , Humanos , Transmisión Sináptica , Dolor , Ganglios Espinales
6.
Brain ; 145(10): 3637-3653, 2022 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-34957475

RESUMEN

Patients with bi-allelic loss of function mutations in the voltage-gated sodium channel Nav1.7 present with congenital insensitivity to pain (CIP), whilst low threshold mechanosensation is reportedly normal. Using psychophysics (n = 6 CIP participants and n = 86 healthy controls) and facial electromyography (n = 3 CIP participants and n = 8 healthy controls), we found that these patients also have abnormalities in the encoding of affective touch, which is mediated by the specialized afferents C-low threshold mechanoreceptors (C-LTMRs). In the mouse, we found that C-LTMRs express high levels of Nav1.7. Genetic loss or selective pharmacological inhibition of Nav1.7 in C-LTMRs resulted in a significant reduction in the total sodium current density, an increased mechanical threshold and reduced sensitivity to non-noxious cooling. The behavioural consequence of loss of Nav1.7 in C-LTMRs in mice was an elevation in the von Frey mechanical threshold and less sensitivity to cooling on a thermal gradient. Nav1.7 is therefore not only essential for normal pain perception but also for normal C-LTMR function, cool sensitivity and affective touch.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7 , Insensibilidad Congénita al Dolor , Animales , Humanos , Ratones , Mecanorreceptores , Canal de Sodio Activado por Voltaje NAV1.7/genética , Insensibilidad Congénita al Dolor/genética , Sodio
7.
J Neurosci ; 41(39): 8249-8261, 2021 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-34400519

RESUMEN

Pain is the major debilitating symptom of osteoarthritis (OA), which is difficult to treat. In OA patients joint tissue damage only poorly associates with pain, indicating other mechanisms contribute to OA pain. Immune cells regulate the sensory system, but little is known about the involvement of immune cells in OA pain. Here, we report that macrophages accumulate in the dorsal root ganglia (DRG) distant from the site of injury in two rodent models of OA. DRG macrophages acquired an M1-like phenotype, and depletion of DRG macrophages resolved OA pain in male and female mice. Sensory neurons innervating the damaged knee joint shape DRG macrophages into an M1-like phenotype. Persisting OA pain, accumulation of DRG macrophages, and programming of DRG macrophages into an M1-like phenotype were independent of Nav1.8 nociceptors. Inhibition of M1-like macrophages in the DRG by intrathecal injection of an IL4-IL10 fusion protein or M2-like macrophages resolved persistent OA pain. In conclusion, these findings reveal a crucial role for macrophages in maintaining OA pain independent of the joint damage and suggest a new direction to treat OA pain.SIGNIFICANCE STATEMENT In OA patients pain poorly correlates with joint tissue changes indicating mechanisms other than only tissue damage that cause pain in OA. We identified that DRG containing the somata of sensory neurons innervating the damaged knee are infiltrated with macrophages that are shaped into an M1-like phenotype by sensory neurons. We show that these DRG macrophages actively maintain OA pain remotely and independent of joint damage. The phenotype of these macrophages is crucial for a pain-promoting role. Targeting the phenotype of DRG macrophages with either M2-like macrophages or a cytokine fusion protein that skews macrophages into an M2-like phenotype resolves OA pain. Our work reveals a mechanism that contributes to the maintenance of OA pain distant from the affected knee joint and suggests that dorsal root ganglia macrophages are a target to treat osteoarthritis chronic pain.


Asunto(s)
Artritis Experimental/metabolismo , Ganglios Espinales/metabolismo , Macrófagos/metabolismo , Osteoartritis/metabolismo , Dolor/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Femenino , Masculino , Ratones , Nociceptores/fisiología
8.
EMBO J ; 37(3): 427-445, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29335280

RESUMEN

The voltage-gated sodium channel NaV1.7 plays a critical role in pain pathways. We generated an epitope-tagged NaV1.7 mouse that showed normal pain behaviours to identify channel-interacting proteins. Analysis of NaV1.7 complexes affinity-purified under native conditions by mass spectrometry revealed 267 proteins associated with Nav1.7 in vivo The sodium channel ß3 (Scn3b), rather than the ß1 subunit, complexes with Nav1.7, and we demonstrate an interaction between collapsing-response mediator protein (Crmp2) and Nav1.7, through which the analgesic drug lacosamide regulates Nav1.7 current density. Novel NaV1.7 protein interactors including membrane-trafficking protein synaptotagmin-2 (Syt2), L-type amino acid transporter 1 (Lat1) and transmembrane P24-trafficking protein 10 (Tmed10) together with Scn3b and Crmp2 were validated by co-immunoprecipitation (Co-IP) from sensory neuron extract. Nav1.7, known to regulate opioid receptor efficacy, interacts with the G protein-regulated inducer of neurite outgrowth (Gprin1), an opioid receptor-binding protein, demonstrating a physical and functional link between Nav1.7 and opioid signalling. Further information on physiological interactions provided with this normal epitope-tagged mouse should provide useful insights into the many functions now associated with the NaV1.7 channel.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Dolor/fisiopatología , Receptores de N-Metil-D-Aspartato/metabolismo , Receptores Opioides/metabolismo , Células Receptoras Sensoriales/metabolismo , Acetamidas/farmacología , Analgésicos/farmacología , Animales , Línea Celular , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Lacosamida , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Canal de Sodio Activado por Voltaje NAV1.7/genética , Unión Proteica , Mapeo de Interacción de Proteínas , Transporte de Proteínas/fisiología , Sinaptotagmina II/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Subunidad beta-3 de Canal de Sodio Activado por Voltaje/metabolismo
9.
Brain ; 144(6): 1711-1726, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-33693512

RESUMEN

Patients with neuropathic pain often experience innocuous cooling as excruciating pain. The cell and molecular basis of this cold allodynia is little understood. We used in vivo calcium imaging of sensory ganglia to investigate how the activity of peripheral cold-sensing neurons was altered in three mouse models of neuropathic pain: oxaliplatin-induced neuropathy, partial sciatic nerve ligation, and ciguatera poisoning. In control mice, cold-sensing neurons were few in number and small in size. In neuropathic animals with cold allodynia, a set of normally silent large diameter neurons became sensitive to cooling. Many of these silent cold-sensing neurons responded to noxious mechanical stimuli and expressed the nociceptor markers Nav1.8 and CGRPα. Ablating neurons expressing Nav1.8 resulted in diminished cold allodynia. The silent cold-sensing neurons could also be activated by cooling in control mice through blockade of Kv1 voltage-gated potassium channels. Thus, silent cold-sensing neurons are unmasked in diverse neuropathic pain states and cold allodynia results from peripheral sensitization caused by altered nociceptor excitability.


Asunto(s)
Frío/efectos adversos , Hiperalgesia/metabolismo , Neuralgia/metabolismo , Neuronas/metabolismo , Nociceptores/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Receptores de Péptido Relacionado con el Gen de Calcitonina/metabolismo , Canales de Potasio de la Superfamilia Shaker/metabolismo , Sensación Térmica/fisiología
10.
Annu Rev Pharmacol Toxicol ; 58: 123-142, 2018 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-28968191

RESUMEN

Pain is an increasing clinical challenge affecting about half the population, with a substantial number of people suffering daily intense pain. Such suffering can be linked to the dramatic rise in opioid use and associated deaths in the United States. There is a pressing need for new analgesics with limited side effects. Here, we summarize what we know about the genetics of pain and implications for drug development. We make the case that chronic pain is not one but a set of disease states, with peripheral drive a key element in most. We argue that understanding redundancy and plasticity, hallmarks of the nervous system, is critical in developing analgesic drug strategies. We describe the exploitation of monogenic pain syndromes and genetic association studies to define analgesic targets, as well as issues associated with animal models of pain. We appraise present-day screening technologies and describe recent approaches to pain treatment that hold promise.


Asunto(s)
Analgésicos/uso terapéutico , Dolor Crónico/tratamiento farmacológico , Dolor Crónico/genética , Animales , Desarrollo de Medicamentos/métodos , Genética , Humanos , Estados Unidos
11.
Annu Rev Neurosci ; 36: 519-46, 2013 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-23750514

RESUMEN

Transient receptor potential (TRP) channels act as sensors for a range of stimuli as diverse as light, sound, touch, pheromones, and tissue damage. Their role in mechanosensation in the animal kingdom, identified by gene ablation studies, has raised questions about whether they are directly mechanically gated, whether they act alone or in concert with other channels to transduce mechanical stimuli, and their relative importance in various functions and disease states in humans. The ability of these channels to form heteromultimers and interact with other ion channels underlies a range of cell-specific functions in different cell types. Here we overview recent advances in this rapidly expanding field, focusing on somatosensation, hearing, the cardiovascular system, and interactions between TRP channels and other proteins involved in mechanoelectrical signaling.


Asunto(s)
Mecanorreceptores/fisiología , Mecanotransducción Celular/fisiología , Tacto/fisiología , Canales de Potencial de Receptor Transitorio/fisiología , Animales , Humanos
12.
Nature ; 510(7503): 157-61, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24759321

RESUMEN

The skin has a dual function as a barrier and a sensory interface between the body and the environment. To protect against invading pathogens, the skin harbours specialized immune cells, including dermal dendritic cells (DDCs) and interleukin (IL)-17-producing γδ T (γδT17) cells, the aberrant activation of which by IL-23 can provoke psoriasis-like inflammation. The skin is also innervated by a meshwork of peripheral nerves consisting of relatively sparse autonomic and abundant sensory fibres. Interactions between the autonomic nervous system and immune cells in lymphoid organs are known to contribute to systemic immunity, but how peripheral nerves regulate cutaneous immune responses remains unclear. We exposed the skin of mice to imiquimod, which induces IL-23-dependent psoriasis-like inflammation. Here we show that a subset of sensory neurons expressing the ion channels TRPV1 and Nav1.8 is essential to drive this inflammatory response. Imaging of intact skin revealed that a large fraction of DDCs, the principal source of IL-23, is in close contact with these nociceptors. Upon selective pharmacological or genetic ablation of nociceptors, DDCs failed to produce IL-23 in imiquimod-exposed skin. Consequently, the local production of IL-23-dependent inflammatory cytokines by dermal γδT17 cells and the subsequent recruitment of inflammatory cells to the skin were markedly reduced. Intradermal injection of IL-23 bypassed the requirement for nociceptor communication with DDCs and restored the inflammatory response. These findings indicate that TRPV1(+)Nav1.8(+) nociceptors, by interacting with DDCs, regulate the IL-23/IL-17 pathway and control cutaneous immune responses.


Asunto(s)
Interleucina-23/inmunología , Nociceptores/metabolismo , Psoriasis/inmunología , Psoriasis/patología , Células Receptoras Sensoriales/metabolismo , Piel/inervación , Piel/patología , Aminoquinolinas , Animales , Modelos Animales de Enfermedad , Femenino , Imiquimod , Inflamación/inducido químicamente , Inflamación/inmunología , Inflamación/patología , Interleucina-17/biosíntesis , Interleucina-17/inmunología , Interleucina-23/biosíntesis , Interleucinas/biosíntesis , Interleucinas/inmunología , Células de Langerhans/inmunología , Células de Langerhans/metabolismo , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Canal de Sodio Activado por Voltaje NAV1.8/metabolismo , Nociceptores/efectos de los fármacos , Psoriasis/inducido químicamente , Células Receptoras Sensoriales/efectos de los fármacos , Piel/citología , Piel/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Canales Catiónicos TRPV/metabolismo , Interleucina-22
13.
Nature ; 516(7529): 121-5, 2014 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-25471886

RESUMEN

The sense of touch provides critical information about our physical environment by transforming mechanical energy into electrical signals. It is postulated that mechanically activated cation channels initiate touch sensation, but the identity of these molecules in mammals has been elusive. Piezo2 is a rapidly adapting, mechanically activated ion channel expressed in a subset of sensory neurons of the dorsal root ganglion and in cutaneous mechanoreceptors known as Merkel-cell-neurite complexes. It has been demonstrated that Merkel cells have a role in vertebrate mechanosensation using Piezo2, particularly in shaping the type of current sent by the innervating sensory neuron; however, major aspects of touch sensation remain intact without Merkel cell activity. Here we show that mice lacking Piezo2 in both adult sensory neurons and Merkel cells exhibit a profound loss of touch sensation. We precisely localize Piezo2 to the peripheral endings of a broad range of low-threshold mechanoreceptors that innervate both hairy and glabrous skin. Most rapidly adapting, mechanically activated currents in dorsal root ganglion neuronal cultures are absent in Piezo2 conditional knockout mice, and ex vivo skin nerve preparation studies show that the mechanosensitivity of low-threshold mechanoreceptors strongly depends on Piezo2. This cellular phenotype correlates with an unprecedented behavioural phenotype: an almost complete deficit in light-touch sensation in multiple behavioural assays, without affecting other somatosensory functions. Our results highlight that a single ion channel that displays rapidly adapting, mechanically activated currents in vitro is responsible for the mechanosensitivity of most low-threshold mechanoreceptor subtypes involved in innocuous touch sensation. Notably, we find that touch and pain sensation are separable, suggesting that as-yet-unknown mechanically activated ion channel(s) must account for noxious (painful) mechanosensation.


Asunto(s)
Canales Iónicos/metabolismo , Mecanotransducción Celular/fisiología , Piel/inervación , Tacto/fisiología , Animales , Canales Iónicos/genética , Mecanorreceptores/metabolismo , Mecanotransducción Celular/genética , Células de Merkel/fisiología , Ratones , Ratones Noqueados , Células Receptoras Sensoriales/fisiología , Tacto/genética
14.
Brain ; 141(4): 1028-1039, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29394316

RESUMEN

Many studies support the pro-nociceptive role of brain-derived neurotrophin factor (BDNF) in pain processes in the peripheral and central nervous system. We have previously shown that nociceptor-derived BDNF is involved in inflammatory pain. Microglial-derived BDNF has also been shown to be involved in neuropathic pain. However, the distinct contribution of primary afferent-derived BNDF to chronic pain processing remains undetermined. In this study, we used Avil-CreERT2 mice to delete Bdnf from all adult peripheral sensory neurons. Conditional BDNF knockouts were healthy with no sensory neuron loss. Behavioural assays and in vivo electrophysiology indicated that spinal excitability was normal. Following formalin inflammation or neuropathy with a modified Chung model, we observed normal development of acute pain behaviour, but a deficit in second phase formalin-induced nocifensive responses and a reversal of neuropathy-induced mechanical hypersensitivity during the later chronic pain phase in conditional BDNF knockout mice. In contrast, we observed normal development of acute and chronic neuropathic pain in the Seltzer model, indicating differences in the contribution of BDNF to distinct models of neuropathy. We further used a model of hyperalgesic priming to examine the contribution of primary afferent-derived BDNF in the transition from acute to chronic pain, and found that primed BDNF knockout mice do not develop prolonged mechanical hypersensitivity to an inflammatory insult. Our data suggest that BDNF derived from sensory neurons plays a critical role in mediating the transition from acute to chronic pain.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/metabolismo , Dolor Crónico/patología , Ganglios Espinales/patología , Células Receptoras Sensoriales/metabolismo , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Carragenina/toxicidad , Dolor Crónico/inducido químicamente , Modelos Animales de Enfermedad , Femenino , Formaldehído/toxicidad , Hiperalgesia/etiología , Masculino , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Dimensión del Dolor
15.
Brain ; 141(2): 365-376, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29253101

RESUMEN

Chronic pain is a major global public health issue causing a severe impact on both the quality of life for sufferers and the wider economy. Despite the significant clinical burden, little progress has been made in terms of therapeutic development. A unique approach to identifying new human-validated analgesic drug targets is to study rare families with inherited pain insensitivity. Here we have analysed an otherwise normal family where six affected individuals display a pain insensitive phenotype that is characterized by hyposensitivity to noxious heat and painless bone fractures. This autosomal dominant disorder is found in three generations and is not associated with a peripheral neuropathy. A novel point mutation in ZFHX2, encoding a putative transcription factor expressed in small diameter sensory neurons, was identified by whole exome sequencing that segregates with the pain insensitivity. The mutation is predicted to change an evolutionarily highly conserved arginine residue 1913 to a lysine within a homeodomain. Bacterial artificial chromosome (BAC) transgenic mice bearing the orthologous murine p.R1907K mutation, as well as Zfhx2 null mutant mice, have significant deficits in pain sensitivity. Gene expression analyses in dorsal root ganglia from mutant and wild-type mice show altered expression of genes implicated in peripheral pain mechanisms. The ZFHX2 variant and downstream regulated genes associated with a human pain-insensitive phenotype are therefore potential novel targets for the development of new analgesic drugs.awx326media15680039660001.


Asunto(s)
Insensibilidad Congénita al Dolor/genética , Umbral del Dolor/fisiología , Dolor/fisiopatología , Mutación Puntual/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Adolescente , Adulto , Anciano , Animales , Calcio/metabolismo , Capsaicina/efectos adversos , Modelos Animales de Enfermedad , Femenino , Ganglios Espinales/patología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Humanos , Hiperalgesia/patología , Hiperalgesia/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Dolor/inducido químicamente , Insensibilidad Congénita al Dolor/patología , Insensibilidad Congénita al Dolor/fisiopatología , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/fisiología , Piel/patología , Adulto Joven
16.
J Physiol ; 595(8): 2661-2679, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28105664

RESUMEN

KEY POINTS: Voltage-gated sodium channels play a fundamental role in determining neuronal excitability. Specifically, voltage-gated sodium channel subtype NaV 1.7 is required for sensing acute and inflammatory somatic pain in mice and humans but its significance in pain originating from the viscera is unknown. Using comparative behavioural models evoking somatic and visceral pain pathways, we identify the requirement for NaV 1.7 in regulating somatic (noxious heat pain threshold) but not in visceral pain signalling. These results enable us to better understand the mechanisms underlying the transduction of noxious stimuli from the viscera, suggest that the investigation of pain pathways should be undertaken in a modality-specific manner and help to direct drug discovery efforts towards novel visceral analgesics. ABSTRACT: Voltage-gated sodium channel NaV 1.7 is required for acute and inflammatory pain in mice and humans but its significance for visceral pain is unknown. Here we examine the role of NaV 1.7 in visceral pain processing and the development of referred hyperalgesia using a conditional nociceptor-specific NaV 1.7 knockout mouse (NaV 1.7Nav1.8 ) and selective small-molecule NaV 1.7 antagonist PF-5198007. NaV 1.7Nav1.8 mice showed normal nociceptive behaviours in response to intracolonic application of either capsaicin or mustard oil, stimuli known to evoke sustained nociceptor activity and sensitization following tissue damage, respectively. Normal responses following induction of cystitis by cyclophosphamide were also observed in both NaV 1.7Nav1.8 and littermate controls. Loss, or blockade, of NaV 1.7 did not affect afferent responses to noxious mechanical and chemical stimuli in nerve-gut preparations in mouse, or following antagonism of NaV 1.7 in resected human appendix stimulated by noxious distending pressures. However, expression analysis of voltage-gated sodium channel α subunits revealed NaV 1.7 mRNA transcripts in nearly all retrogradely labelled colonic neurons, suggesting redundancy in function. By contrast, using comparative somatic behavioural models we identify that genetic deletion of NaV 1.7 (in NaV 1.8-expressing neurons) regulates noxious heat pain threshold and that this can be recapitulated by the selective NaV 1.7 antagonist PF-5198007. Our data demonstrate that NaV 1.7 (in NaV 1.8-expressing neurons) contributes to defined pain pathways in a modality-dependent manner, modulating somatic noxious heat pain, but is not required for visceral pain processing, and advocate that pharmacological block of NaV 1.7 alone in the viscera may be insufficient in targeting chronic visceral pain.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/deficiencia , Nociceptores/metabolismo , Dolor Visceral/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Animales , Capsaicina/toxicidad , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Planta de la Mostaza/toxicidad , Canal de Sodio Activado por Voltaje NAV1.7/genética , Dolor Nociceptivo/inducido químicamente , Dolor Nociceptivo/genética , Dolor Nociceptivo/metabolismo , Nociceptores/efectos de los fármacos , Aceites de Plantas/toxicidad , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Bloqueadores de los Canales de Sodio/farmacología , Dolor Visceral/inducido químicamente , Dolor Visceral/genética
17.
Nature ; 472(7342): 186-90, 2011 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-21441906

RESUMEN

Loss of function of the gene SCN9A, encoding the voltage-gated sodium channel Na(v)1.7, causes a congenital inability to experience pain in humans. Here we show that Na(v)1.7 is not only necessary for pain sensation but is also an essential requirement for odour perception in both mice and humans. We examined human patients with loss-of-function mutations in SCN9A and show that they are unable to sense odours. To establish the essential role of Na(v)1.7 in odour perception, we generated conditional null mice in which Na(v)1.7 was removed from all olfactory sensory neurons. In the absence of Na(v)1.7, these neurons still produce odour-evoked action potentials but fail to initiate synaptic signalling from their axon terminals at the first synapse in the olfactory system. The mutant mice no longer display vital, odour-guided behaviours such as innate odour recognition and avoidance, short-term odour learning, and maternal pup retrieval. Our study creates a mouse model of congenital general anosmia and provides new strategies to explore the genetic basis of the human sense of smell.


Asunto(s)
Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutación/genética , Trastornos del Olfato/genética , Trastornos del Olfato/fisiopatología , Canales de Sodio/genética , Potenciales de Acción , Animales , Conducta Animal , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Canal de Sodio Activado por Voltaje NAV1.7 , Odorantes/análisis , Trastornos del Olfato/congénito , Trastornos del Olfato/patología , Mucosa Olfatoria/citología , Mucosa Olfatoria/patología , Vías Olfatorias/metabolismo , Vías Olfatorias/patología , Vías Olfatorias/fisiopatología , Percepción Olfatoria/genética , Percepción Olfatoria/fisiología , Neuronas Receptoras Olfatorias/metabolismo , Neuronas Receptoras Olfatorias/patología , Dolor/genética , Dolor/fisiopatología , Fenotipo , Olfato/genética , Olfato/fisiología , Canales de Sodio/deficiencia , Canales de Sodio/metabolismo , Sinapsis/metabolismo , Sinapsis/patología , Orina/química
18.
Mol Cell Neurosci ; 75: 36-43, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27346077

RESUMEN

BACKGROUND: MicroRNAs (miRNAs) regulate gene expression in physiological as well as in pathological processes, including chronic pain. Whether deletion of a gene can affect expression of the miRNAs that associate with the deleted gene mRNA remains elusive. We investigated the effects of brain-derived neurotrophic factor (Bdnf) gene deletion on the expression of miR-1 in dorsal root ganglion (DRG) neurons and its pain-associated downstream targets heat shock protein 60 (Hsp60) and connexin 43 (Cx43) in tamoxifen-inducible conditional knockout mice, Bdnf(fl/fl); Advillin-CreER(T2) (Bdnf cKO). RESULTS: Efficient Bdnf gene deletion was confirmed in DRG of Bdnf cKO mice by Real-Time qRT-PCR and ELISA 10days after completed tamoxifen treatment. In DRG, miR-1 expression was reduced 0.44-fold (p<0.05; Real-time qRT-PCR) in Bdnf cKO compared to floxed wildtype littermate control Bdnf(fl/fl) mice (WT). While Hsp60 protein expression was increased 1.85-fold (p<0.05; Western blot analysis), expression levels of Cx43 and the miR-1-associated transcription factors MEF2a and SRF remained unchanged. When analyzing Bdnf cKO mice 32days after complete tamoxifen treatment to investigate whether observed expression alterations remain permanently, we found no significant differences between Bdnf cKO and WT mice. However, miRNA microarray analysis revealed that 167 miRNAs altered (p<0.05) in DRG of these mice following Bdnf gene deletion. CONCLUSIONS: Our results indicate that deletion of Bdnf in DRG neurons leads to a temporary dysregulation of miR-1, suggesting an impairment of a presumable feedback loop between BDNF protein and its targeting miR-1. This appears to affect its downstream protein Hsp60 and as a consequence might influence the phenotype after inducible Bdnf gene deletion. While this appears to be a MEF2a-/SRF-independent and transient effect, expression levels of various other miRNAs may remain permanently altered.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo/genética , Ganglios Espinales/metabolismo , MicroARNs/genética , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Chaperonina 60/genética , Chaperonina 60/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Femenino , Eliminación de Gen , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo
19.
Mar Drugs ; 15(6)2017 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-28635651

RESUMEN

Visceral pain is very common and represents a major unmet clinical need for which current pharmacological treatments are often insufficient. Tetrodotoxin (TTX) is a potent neurotoxin that exerts analgesic actions in both humans and rodents under different somatic pain conditions, but its effect has been unexplored in visceral pain. Therefore, we tested the effects of systemic TTX in viscero-specific mouse models of chemical stimulation of the colon (intracolonic instillation of capsaicin and mustard oil) and intraperitoneal cyclophosphamide-induced cystitis. The subcutaneous administration of TTX dose-dependently inhibited the number of pain-related behaviors in all evaluated pain models and reversed the referred mechanical hyperalgesia (examined by stimulation of the abdomen with von Frey filaments) induced by capsaicin and cyclophosphamide, but not that induced by mustard oil. Morphine inhibited both pain responses and the referred mechanical hyperalgesia in all tests. Conditional nociceptor­specific Nav1.7 knockout mice treated with TTX showed the same responses as littermate controls after the administration of the algogens. No motor incoordination after the administration of TTX was observed. These results suggest that blockade of TTX-sensitive sodium channels, but not Nav1.7 subtype alone, by systemic administration of TTX might be a potential therapeutic strategy for the treatment of visceral pain.


Asunto(s)
Dimensión del Dolor/efectos de los fármacos , Tetrodotoxina/farmacología , Dolor Visceral/tratamiento farmacológico , Analgésicos/farmacología , Animales , Capsaicina/farmacología , Colon/efectos de los fármacos , Colon/metabolismo , Cistitis/tratamiento farmacológico , Cistitis/metabolismo , Modelos Animales de Enfermedad , Femenino , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Masculino , Ratones , Ratones Noqueados , Morfina/farmacología , Planta de la Mostaza , Nociceptores/metabolismo , Aceites de Plantas/farmacología , Canales de Sodio/metabolismo , Dolor Visceral/metabolismo
20.
EMBO J ; 31(19): 3795-808, 2012 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-22850668

RESUMEN

Ciguatoxins are sodium channel activator toxins that cause ciguatera, the most common form of ichthyosarcotoxism, which presents with peripheral sensory disturbances, including the pathognomonic symptom of cold allodynia which is characterized by intense stabbing and burning pain in response to mild cooling. We show that intraplantar injection of P-CTX-1 elicits cold allodynia in mice by targeting specific unmyelinated and myelinated primary sensory neurons. These include both tetrodotoxin-resistant, TRPA1-expressing peptidergic C-fibres and tetrodotoxin-sensitive A-fibres. P-CTX-1 does not directly open heterologously expressed TRPA1, but when co-expressed with Na(v) channels, sodium channel activation by P-CTX-1 is sufficient to drive TRPA1-dependent calcium influx that is responsible for the development of cold allodynia, as evidenced by a large reduction of excitatory effect of P-CTX-1 on TRPA1-deficient nociceptive C-fibres and of ciguatoxin-induced cold allodynia in TRPA1-null mutant mice. Functional MRI studies revealed that ciguatoxin-induced cold allodynia enhanced the BOLD (Blood Oxygenation Level Dependent) signal, an effect that was blunted in TRPA1-deficient mice, confirming an important role for TRPA1 in the pathogenesis of cold allodynia.


Asunto(s)
Ciguatoxinas/toxicidad , Dolor/inducido químicamente , Animales , Frío , Hiperalgesia/inducido químicamente , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Ratas Wistar , Células Receptoras Sensoriales/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio/efectos de los fármacos , Canales de Potencial de Receptor Transitorio/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA