Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 170(1): 185-198.e16, 2017 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-28648659

RESUMEN

Dietary, microbial, and inflammatory factors modulate the gut-brain axis and influence physiological processes ranging from metabolism to cognition. The gut epithelium is a principal site for detecting such agents, but precisely how it communicates with neural elements is poorly understood. Serotonergic enterochromaffin (EC) cells are proposed to fulfill this role by acting as chemosensors, but understanding how these rare and unique cell types transduce chemosensory information to the nervous system has been hampered by their paucity and inaccessibility to single-cell measurements. Here, we circumvent this limitation by exploiting cultured intestinal organoids together with single-cell measurements to elucidate intrinsic biophysical, pharmacological, and genetic properties of EC cells. We show that EC cells express specific chemosensory receptors, are electrically excitable, and modulate serotonin-sensitive primary afferent nerve fibers via synaptic connections, enabling them to detect and transduce environmental, metabolic, and homeostatic information from the gut directly to the nervous system.


Asunto(s)
Células Quimiorreceptoras/metabolismo , Células Enterocromafines/metabolismo , Tracto Gastrointestinal/citología , Vías Nerviosas , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Canales de Calcio/metabolismo , Catecolaminas/metabolismo , Perfilación de la Expresión Génica , Humanos , Síndrome del Colon Irritable/patología , Ratones , Fibras Nerviosas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores Odorantes/metabolismo , Receptores de Serotonina 5-HT3/metabolismo , Serotonina/metabolismo , Transducción de Señal , Sinapsis/metabolismo , Canal Catiónico TRPA1 , Canales de Potencial de Receptor Transitorio/metabolismo
2.
Nature ; 616(7955): 137-142, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36949192

RESUMEN

Gastrointestinal (GI) discomfort is a hallmark of most gut disorders and represents an important component of chronic visceral pain1. For the growing population afflicted by irritable bowel syndrome, GI hypersensitivity and pain persist long after tissue injury has resolved2. Irritable bowel syndrome also exhibits a strong sex bias, afflicting women three times more than men1. Here, we focus on enterochromaffin (EC) cells, which are rare excitable, serotonergic neuroendocrine cells in the gut epithelium3-5. EC cells detect and transduce noxious stimuli to nearby mucosal nerve endings3,6 but involvement of this signalling pathway in visceral pain and attendant sex differences has not been assessed. By enhancing or suppressing EC cell function in vivo, we show that these cells are sufficient to elicit hypersensitivity to gut distension and necessary for the sensitizing actions of isovalerate, a bacterial short-chain fatty acid associated with GI inflammation7,8. Remarkably, prolonged EC cell activation produced persistent visceral hypersensitivity, even in the absence of an instigating inflammatory episode. Furthermore, perturbing EC cell activity promoted anxiety-like behaviours which normalized after blockade of serotonergic signalling. Sex differences were noted across a range of paradigms, indicating that the EC cell-mucosal afferent circuit is tonically engaged in females. Our findings validate a critical role for EC cell-mucosal afferent signalling in acute and persistent GI pain, in addition to highlighting genetic models for studying visceral hypersensitivity and the sex bias of gut pain.


Asunto(s)
Ansiedad , Células Enterocromafines , Dolor Visceral , Femenino , Humanos , Masculino , Ansiedad/complicaciones , Ansiedad/fisiopatología , Sistema Digestivo/inervación , Sistema Digestivo/fisiopatología , Células Enterocromafines/metabolismo , Síndrome del Colon Irritable/complicaciones , Síndrome del Colon Irritable/fisiopatología , Síndrome del Colon Irritable/psicología , Caracteres Sexuales , Dolor Visceral/complicaciones , Dolor Visceral/fisiopatología , Dolor Visceral/psicología , Inflamación/complicaciones , Inflamación/fisiopatología , Serotonina/metabolismo , Reproducibilidad de los Resultados
3.
J Neurochem ; 2023 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-37165846

RESUMEN

Chronic pelvic pain (CPP) is the most debilitating symptom of gynaecological disorders such as endometriosis. However, it remains unclear how sensory neurons from pelvic organs affected by endometriosis, such as the female reproductive tract, detect and transmit nociceptive events and how these signals are processed within the central nervous system (CNS). Using a previously characterized mouse model of endometriosis, we investigated whether the increased pain sensitivity occurring in endometriosis could be attributed to (i) changes in mechanosensory properties of sensory afferents innervating the reproductive tract, (ii) alterations in sensory input from reproductive organs to the spinal cord or (iii) neuroinflammation and sensitization of spinal neural circuits. Mechanosensitivity of vagina-innervating primary afferents was examined using an ex vivo single-unit extracellular recording preparation. Nociceptive signalling from the vagina to the spinal cord was quantified by phosphorylated MAP kinase ERK1/2 immunoreactivity. Immunohistochemistry was used to determine glial and neuronal circuit alterations within the spinal cord. We found that sensory afferents innervating the rostral, but not caudal portions of the mouse vagina, developed mechanical hypersensitivity in endometriosis. Nociceptive signalling from the vagina to the spinal cord was significantly enhanced in mice with endometriosis. Moreover, mice with endometriosis developed microgliosis, astrogliosis and enhanced substance P neurokinin-1 receptor immunoreactivity within the spinal cord, suggesting the development of neuroinflammation and sensitization of spinal circuitry in endometriosis. These results demonstrate endometriosis-induced neuroplasticity occurring at both peripheral and central sites of sensory afferent pathways. These findings may help to explain the altered sensitivity to pain in endometriosis and provide a novel platform for targeted pain relief treatments for this debilitating disorder.

4.
J Neurochem ; 2023 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-36840383

RESUMEN

Chronic pelvic pain (CPP) is the primary symptom of endometriosis patients, but adequate treatments are lacking. Modulation of ion channels expressed by sensory nerves innervating the viscera has shown promise for the treatment of irritable bowel syndrome and overactive bladder. However, similar approaches for endometriosis-associated CPP remain underdeveloped. Here, we examined the role of the voltage-gated sodium (NaV ) channel NaV 1.7 in (i) the sensitivity of vagina-innervating sensory afferents and investigated whether (ii) NaV 1.7 inhibition reduces nociceptive signals from the vagina and (iii) ameliorates endometriosis-associated CPP. The mechanical responsiveness of vagina-innervating sensory afferents was assessed with ex vivo single-unit recording preparations. Pain evoked by vaginal distension (VD) was quantified by the visceromotor response (VMR) in vivo. In control mice, pharmacological activation of NaV 1.7 with OD1 sensitised vagina-innervating pelvic afferents to mechanical stimuli. Using a syngeneic mouse model of endometriosis, we established that endometriosis sensitised vagina-innervating pelvic afferents to mechanical stimuli. The highly selective NaV 1.7 inhibitor Tsp1a revealed that this afferent hypersensitivity occurred in a NaV 1.7-dependent manner. Moreover, in vivo intra-vaginal treatment with Tsp1a reduced the exaggerated VMRs to VD which is characteristic of mice with endometriosis. Conversely, Tsp1a did not alter ex vivo afferent mechanosensitivity nor in vivo VMRs to VD in Sham control mice. Collectively, these findings suggest that NaV 1.7 plays a crucial role in endometriosis-induced vaginal hyperalgesia. Importantly, NaV 1.7 inhibition selectively alleviated endometriosis-associated CPP without the loss of normal sensation, suggesting that selective targeting of NaV 1.7 could improve the quality of life of women with endometriosis.

5.
J Neurosci ; 41(17): 3900-3916, 2021 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-33727332

RESUMEN

Understanding the sensory mechanisms innervating the bladder is paramount to developing efficacious treatments for chronic bladder hypersensitivity conditions. The contribution of Mas-gene-related G protein-coupled receptors (Mrgpr) to bladder signaling is currently unknown. Using male and female mice, we show with single-cell RT-PCR that subpopulations of DRG neurons innervating the mouse bladder express MrgprA3 (14%) and MrgprC11 (38%), either individually or in combination, with high levels of coexpression with Trpv1 (81%-89%). Calcium imaging studies demonstrated MrgprA3 and MrgprC11 agonists (chloroquine, BAM8-22, and neuropeptide FF) activated subpopulations of bladder-innervating DRG neurons, showing functional evidence of coexpression between MrgprA3, MrgprC11, and TRPV1. In ex vivo bladder-nerve preparations, chloroquine, BAM8-22, and neuropeptide FF all evoked mechanical hypersensitivity in subpopulations (20%-41%) of bladder afferents. These effects were absent in recordings from Mrgpr-clusterΔ-/- mice. In vitro whole-cell patch-clamp recordings showed that application of an MrgprA3/C11 agonist mixture induced neuronal hyperexcitability in 44% of bladder-innervating DRG neurons. Finally, in vivo instillation of an MrgprA3/C11 agonist mixture into the bladder of WT mice induced a significant activation of dorsal horn neurons within the lumbosacral spinal cord, as quantified by pERK immunoreactivity. This MrgprA3/C11 agonist-induced activation was particularly apparent within the superficial dorsal horn and the sacral parasympathetic nuclei of WT, but not Mrgpr-clusterΔ-/- mice. This study demonstrates, for the first time, functional expression of MrgprA3 and MrgprC11 in bladder afferents. Activation of these receptors triggers hypersensitivity to distension, a critically valuable factor for therapeutic target development.SIGNIFICANCE STATEMENT Determining how bladder afferents become sensitized is the first step in finding effective treatments for common urological disorders such as overactive bladder and interstitial cystitis/bladder pain syndrome. Here we show that two of the key receptors, MrgprA3 and MrgprC11, that mediate itch from the skin are also expressed on afferents innervating the bladder. Activation of these receptors results in sensitization of bladder afferents, resulting in sensory signals being sent into the spinal cord that prematurely indicate bladder fullness. Targeting bladder afferents expressing MrgprA3 or MrgprC11 and preventing their sensitization may provide a novel approach for treating overactive bladder and interstitial cystitis/bladder pain syndrome.


Asunto(s)
Neuronas Aferentes/fisiología , Receptores Acoplados a Proteínas G/fisiología , Vejiga Urinaria/inervación , Animales , Femenino , Ganglios Espinales/fisiología , Plexo Lumbosacro/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Placa-Clamp , Estimulación Física , Células del Asta Posterior/fisiología , Canales Catiónicos TRPV/fisiología
6.
FASEB J ; 35(4): e21430, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33749885

RESUMEN

Endometriosis is a painful inflammatory disorder affecting ~10% of women of reproductive age. Although chronic pelvic pain (CPP) remains the main symptom of endometriosis patients, adequate treatments for CPP are lacking. Animal models that recapitulate the features and symptoms experienced by women with endometriosis are essential for investigating the etiology of endometriosis, as well as developing new treatments. In this study, we used an autologous mouse model of endometriosis to examine a combination of disease features and symptoms including: a 10 week time course of endometriotic lesion development; the chronic inflammatory environment and development of neuroangiogenesis within lesions; sensory hypersensitivity and altered pain responses to vaginal, colon, bladder, and skin stimulation in conscious animals; and spontaneous animal behavior. We found significant increases in lesion size from week 6 posttransplant. Lesions displayed endometrial glands, stroma, and underwent neuroangiogenesis. Additionally, peritoneal fluid of mice with endometriosis contained known inflammatory mediators and angiogenic factors. Compared to Sham, mice with endometriosis displayed: enhanced sensitivity to pain evoked by (i) vaginal and (ii) colorectal distension, (iii) altered bladder function and increased sensitivity to cutaneous (iv) thermal and (v) mechanical stimuli. The development of endometriosis had no effect on spontaneous behavior. This study describes a comprehensive characterization of a mouse model of endometriosis, recapitulating the clinical features and symptoms experienced by women with endometriosis. Moreover, it delivers the groundwork to investigate the etiology of endometriosis and provides a platform for the development of therapeutical interventions to manage endometriosis-associated CPP.


Asunto(s)
Enfermedades del Colon/etiología , Endometriosis/patología , Enfermedades de la Piel/etiología , Enfermedades de la Vejiga Urinaria/etiología , Enfermedades Vaginales/etiología , Animales , Biomarcadores/sangre , Biomarcadores/metabolismo , Femenino , Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Actividad Motora , Dolor
7.
Nature ; 534(7608): 494-9, 2016 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-27281198

RESUMEN

Voltage-gated sodium (Nav) channels initiate action potentials in most neurons, including primary afferent nerve fibres of the pain pathway. Local anaesthetics block pain through non-specific actions at all Nav channels, but the discovery of selective modulators would facilitate the analysis of individual subtypes of these channels and their contributions to chemical, mechanical, or thermal pain. Here we identify and characterize spider (Heteroscodra maculata) toxins that selectively activate the Nav1.1 subtype, the role of which in nociception and pain has not been elucidated. We use these probes to show that Nav1.1-expressing fibres are modality-specific nociceptors: their activation elicits robust pain behaviours without neurogenic inflammation and produces profound hypersensitivity to mechanical, but not thermal, stimuli. In the gut, high-threshold mechanosensitive fibres also express Nav1.1 and show enhanced toxin sensitivity in a mouse model of irritable bowel syndrome. Together, these findings establish an unexpected role for Nav1.1 channels in regulating the excitability of sensory nerve fibres that mediate mechanical pain.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Nocicepción/efectos de los fármacos , Nociceptores/efectos de los fármacos , Nociceptores/metabolismo , Venenos de Araña/farmacología , Estrés Mecánico , Animales , Modelos Animales de Enfermedad , Femenino , Ganglios Sensoriales/citología , Hiperalgesia/inducido químicamente , Hiperalgesia/metabolismo , Síndrome del Colon Irritable/metabolismo , Masculino , Vaina de Mielina/metabolismo , Canal de Sodio Activado por Voltaje NAV1.1/química , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/metabolismo , Oocitos/metabolismo , Dolor/inducido químicamente , Dolor/metabolismo , Estructura Terciaria de Proteína , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Arañas/química , Especificidad por Sustrato/efectos de los fármacos , Temperatura
8.
Am J Physiol Gastrointest Liver Physiol ; 320(6): G1131-G1141, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33949199

RESUMEN

Chronic abdominal pain is a common clinical condition experienced by patients with irritable bowel syndrome (IBS). A general lack of suitable treatment options for the management of visceral pain is the major contributing factor to the debilitating nature of the disease. Understanding the underlying causes of chronic visceral pain is pivotal to identifying new effective therapies for IBS. This review provides the current evidence, demonstrating that mediators and receptors that induce itch in the skin also act as "gut irritants" in the gastrointestinal tract. Activation of these receptors triggers specific changes in the neuronal excitability of sensory pathways responsible for the transmission of nociceptive information from the periphery to the central nervous system leading to visceral hypersensitivity and visceral pain. Accumulating evidence points to significant roles of irritant mediators and their receptors in visceral hypersensitivity and thus constitutes potential targets for the development of more effective therapeutic options for IBS.


Asunto(s)
Colon/metabolismo , Hiperalgesia/metabolismo , Síndrome del Colon Irritable/metabolismo , Dolor Visceral/metabolismo , Histamina/metabolismo , Humanos , Mastocitos/metabolismo
9.
Proc Natl Acad Sci U S A ; 115(31): E7438-E7447, 2018 07 31.
Artículo en Inglés | MEDLINE | ID: mdl-30012612

RESUMEN

Once activated at the surface of cells, G protein-coupled receptors (GPCRs) redistribute to endosomes, where they can continue to signal. Whether GPCRs in endosomes generate signals that contribute to human disease is unknown. We evaluated endosomal signaling of protease-activated receptor-2 (PAR2), which has been proposed to mediate pain in patients with irritable bowel syndrome (IBS). Trypsin, elastase, and cathepsin S, which are activated in the colonic mucosa of patients with IBS and in experimental animals with colitis, caused persistent PAR2-dependent hyperexcitability of nociceptors, sensitization of colonic afferent neurons to mechanical stimuli, and somatic mechanical allodynia. Inhibitors of clathrin- and dynamin-dependent endocytosis and of mitogen-activated protein kinase kinase-1 prevented trypsin-induced hyperexcitability, sensitization, and allodynia. However, they did not affect elastase- or cathepsin S-induced hyperexcitability, sensitization, or allodynia. Trypsin stimulated endocytosis of PAR2, which signaled from endosomes to activate extracellular signal-regulated kinase. Elastase and cathepsin S did not stimulate endocytosis of PAR2, which signaled from the plasma membrane to activate adenylyl cyclase. Biopsies of colonic mucosa from IBS patients released proteases that induced persistent PAR2-dependent hyperexcitability of nociceptors, and PAR2 association with ß-arrestins, which mediate endocytosis. Conjugation to cholestanol promoted delivery and retention of antagonists in endosomes containing PAR2 A cholestanol-conjugated PAR2 antagonist prevented persistent trypsin- and IBS protease-induced hyperexcitability of nociceptors. The results reveal that PAR2 signaling from endosomes underlies the persistent hyperexcitability of nociceptors that mediates chronic pain of IBS. Endosomally targeted PAR2 antagonists are potential therapies for IBS pain. GPCRs in endosomes transmit signals that contribute to human diseases.


Asunto(s)
Dolor Crónico/etiología , Endosomas/fisiología , Síndrome del Colon Irritable/fisiopatología , Receptor PAR-2/fisiología , Transducción de Señal/fisiología , Animales , Endocitosis , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Nocicepción , Nociceptores/fisiología , Tripsina/farmacología
10.
Am J Physiol Renal Physiol ; 318(2): F298-F314, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31790304

RESUMEN

Interstitial cystitis/bladder pain syndrome (IC/BPS) is a common chronic pelvic disorder with sensory symptoms of urinary urgency, frequency, and pain, indicating a key role for hypersensitivity of bladder-innervating sensory neurons. The inflammatory mast cell mediator histamine has long been implicated in IC/BPS, yet the direct interactions between histamine and bladder afferents remain unclear. In the present study, we show, using a mouse ex vivo bladder afferent preparation, that intravesical histamine enhanced the mechanosensitivity of subpopulations of afferents to bladder distension. Histamine also recruited "silent afferents" that were previously unresponsive to bladder distension. Furthermore, in vivo intravesical histamine enhanced activation of dorsal horn neurons within the lumbosacral spinal cord, indicating increased afferent signaling in the central nervous system. Quantitative RT-PCR revealed significant expression of histamine receptor subtypes (Hrh1-Hrh3) in mouse lumbosacral dorsal root ganglia (DRG), bladder detrusor smooth muscle, mucosa, and isolated urothelial cells. In DRG, Hrh1 was the most abundantly expressed. Acute histamine exposure evoked Ca2+ influx in select populations of DRG neurons but did not elicit calcium transients in isolated primary urothelial cells. Histamine-induced mechanical hypersensitivity ex vivo was abolished in the presence of the histamine H1 receptor antagonist pyrilamine and was not present in preparations from mice lacking transient receptor potential vanilloid 1 (TRPV1). Together, these results indicate that histamine enhances the sensitivity of bladder afferents to distension via interactions with histamine H1 receptor and TRPV1. This hypersensitivity translates to increased sensory input and activation in the spinal cord, which may underlie the symptoms of bladder hypersensitivity and pain experienced in IC/BPS.


Asunto(s)
Cistitis Intersticial/metabolismo , Histamina/administración & dosificación , Hiperalgesia/metabolismo , Mecanorreceptores/efectos de los fármacos , Mecanotransducción Celular/efectos de los fármacos , Receptores Histamínicos H1/efectos de los fármacos , Canales Catiónicos TRPV/metabolismo , Vejiga Urinaria/inervación , Administración Intravesical , Animales , Señalización del Calcio/efectos de los fármacos , Células Cultivadas , Cistitis Intersticial/fisiopatología , Femenino , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Ganglios Espinales/fisiopatología , Hiperalgesia/fisiopatología , Masculino , Mecanorreceptores/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Umbral del Dolor/efectos de los fármacos , Presión , Receptores Histamínicos H1/metabolismo , Canales Catiónicos TRPV/deficiencia , Canales Catiónicos TRPV/genética , Urotelio/efectos de los fármacos , Urotelio/metabolismo
11.
J Physiol ; 597(14): 3751-3768, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31087362

RESUMEN

KEY POINTS: Voltage-gated sodium channels are critical for peripheral sensory neuron transduction and have been implicated in a number of painful and painless disorders. The ß-scorpion toxin, Cn2, is selective for NaV 1.6 in dorsal root ganglion neurons. NaV 1.6 plays an essential role in peripheral sensory neurons, specifically at the distal terminals of mechanosensing fibres innervating the skin and colon. NaV 1.6 activation also leads to enhanced response to mechanical stimulus in vivo. This works highlights the use of toxins in elucidating pain pathways moreover the importance of non-peripherally restricted NaV isoforms in pain generation. ABSTRACT: Peripheral sensory neurons express multiple voltage-gated sodium channels (NaV ) critical for the initiation and propagation of action potentials and transmission of sensory input. Three pore-forming sodium channel isoforms are primarily expressed in the peripheral nervous system (PNS): NaV 1.7, NaV 1.8 and NaV 1.9. These sodium channels have been implicated in painful and painless channelopathies and there has been intense interest in them as potential therapeutic targets in human pain. Emerging evidence suggests NaV 1.6 channels are an important isoform in pain sensing. This study aimed to assess, using pharmacological approaches, the function of NaV 1.6 channels in peripheral sensory neurons. The potent and NaV 1.6 selective ß-scorpion toxin Cn2 was used to assess the effect of NaV 1.6 channel activation in the PNS. The multidisciplinary approach included Ca2+ imaging, whole-cell patch-clamp recordings, skin-nerve and gut-nerve preparations and in vivo behavioural assessment of pain. Cn2 facilitates NaV 1.6 early channel opening, and increased persistent and resurgent currents in large-diameter dorsal root ganglion (DRG) neurons. This promotes enhanced excitatory drive and tonic action potential firing in these neurons. In addition, NaV 1.6 channel activation in the skin and gut leads to increased response to mechanical stimuli. Finally, intra-plantar injection of Cn2 causes mechanical but not thermal allodynia. This study confirms selectivity of Cn2 on NaV 1.6 channels in sensory neurons. Activation of NaV 1.6 channels, in terminals of the skin and viscera, leads to profound changes in neuronal responses to mechanical stimuli. In conclusion, sensory neurons expressing NaV 1.6 are important for the transduction of mechanical information in sensory afferents innervating the skin and viscera.


Asunto(s)
Potenciales de la Membrana/fisiología , Canal de Sodio Activado por Voltaje NAV1.6/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Femenino , Ganglios Espinales/metabolismo , Hiperalgesia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Dolor/metabolismo , Sistema Nervioso Periférico/metabolismo , Piel/metabolismo , Vísceras/metabolismo , Canales de Sodio Activados por Voltaje/metabolismo
12.
Am J Physiol Gastrointest Liver Physiol ; 317(3): G285-G303, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31188624

RESUMEN

The distal colon is innervated by the splanchnic and pelvic nerves, which relay into the thoracolumbar and lumbosacral spinal cord, respectively. Although the peripheral properties of the colonic afferent nerves within these pathways are well studied, their input into the spinal cord remain ill defined. The use of dual retrograde tracing from the colon wall and lumen, in conjunction with in vivo colorectal distension and spinal neuronal activation labeling with phosphorylated MAPK ERK 1/2 (pERK), allowed us to identify thoracolumbar and lumbosacral spinal cord circuits processing colonic afferent input. In the thoracolumbar dorsal horn, central projections of colonic afferents were primarily labeled from the wall of the colon and localized in laminae I and V. In contrast, lumbosacral projections were identified from both lumen and wall tracing, present within various dorsal horn laminae, collateral tracts, and the dorsal gray commissure. Nonnoxious in vivo colorectal distension evoked significant neuronal activation (pERK-immunoreactivity) within the lumbosacral dorsal horn but not in thoracolumbar regions. However, noxious in vivo colorectal distension evoked significant neuronal activation in both the thoracolumbar and lumbosacral dorsal horn, with the distribution of activated neurons correlating to the pattern of traced projections. Dorsal horn neurons activated by colorectal distension were identified as possible populations of projection neurons or excitatory and inhibitory interneurons based on their neurochemistry. Our findings demonstrate how colonic afferents in splanchnic and pelvic pathways differentially relay mechanosensory information into the spinal cord and contribute to the recruitment of spinal cord pathways processing non-noxious and noxious stimuli.NEW & NOTEWORTHY In mice, retrograde tracing from the colon wall and lumen was used to identify unique populations of afferent neurons and central projections within the spinal cord dorsal horn. We show that there are pronounced differences between the spinal cord regions in the distribution pattern of colonic afferent central projections and the pattern of dorsal horn neuron activation evoked by colorectal distension. These findings demonstrate how colonic afferent input influences spinal processing of colonic mechanosensation.


Asunto(s)
Vías Aferentes/fisiología , Colon/inervación , Neuronas Aferentes/fisiología , Células del Asta Posterior/fisiología , Médula Espinal/metabolismo , Animales , Masculino , Ratones Endogámicos C57BL , Médula Espinal/fisiología
13.
J Physiol ; 596(5): 785-807, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29318638

RESUMEN

Chronic visceral pain, altered motility and bladder dysfunction are common, yet poorly managed symptoms of functional and inflammatory disorders of the gastrointestinal and urinary tracts. Recently, numerous human channelopathies of the voltage-gated sodium (NaV ) channel family have been identified, which induce either painful neuropathies, an insensitivity to pain, or alterations in smooth muscle function. The identification of these disorders, in addition to the recent utilisation of genetically modified NaV mice and specific NaV channel modulators, has shed new light on how NaV channels contribute to the function of neuronal and non-neuronal tissues within the gastrointestinal tract and bladder. Here we review the current pre-clinical and clinical evidence to reveal how the nine NaV channel family members (NaV 1.1-NaV 1.9) contribute to abdominal visceral function in normal and disease states.


Asunto(s)
Nocicepción , Dolor Nociceptivo/fisiopatología , Células Receptoras Sensoriales/patología , Vísceras/patología , Canales de Sodio Activados por Voltaje/metabolismo , Animales , Humanos
14.
Gut ; 66(6): 1083-1094, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-26887818

RESUMEN

OBJECTIVE: α-Conotoxin Vc1.1 is a small disulfide-bonded peptide from the venom of the marine cone snail Conus victoriae. Vc1.1 has antinociceptive actions in animal models of neuropathic pain, but its applicability to inhibiting human dorsal root ganglion (DRG) neuroexcitability and reducing chronic visceral pain (CVP) is unknown. DESIGN: We determined the inhibitory actions of Vc1.1 on human DRG neurons and on mouse colonic sensory afferents in healthy and chronic visceral hypersensitivity (CVH) states. In mice, visceral nociception was assessed by neuronal activation within the spinal cord in response to noxious colorectal distension (CRD). Quantitative-reverse-transcription-PCR, single-cell-reverse-transcription-PCR and immunohistochemistry determined γ-aminobutyric acid receptor B (GABABR) and voltage-gated calcium channel (CaV2.2, CaV2.3) expression in human and mouse DRG neurons. RESULTS: Vc1.1 reduced the excitability of human DRG neurons, whereas a synthetic Vc1.1 analogue that is inactive at GABABR did not. Human DRG neurons expressed GABABR and its downstream effector channels CaV2.2 and CaV2.3. Mouse colonic DRG neurons exhibited high GABABR, CaV2.2 and CaV2.3 expression, with upregulation of the CaV2.2 exon-37a variant during CVH. Vc1.1 inhibited mouse colonic afferents ex vivo and nociceptive signalling of noxious CRD into the spinal cord in vivo, with greatest efficacy observed during CVH. A selective GABABR antagonist prevented Vc1.1-induced inhibition, whereas blocking both CaV2.2 and CaV2.3 caused inhibition comparable with Vc1.1 alone. CONCLUSIONS: Vc1.1-mediated activation of GABABR is a novel mechanism for reducing the excitability of human DRG neurons. Vc1.1-induced activation of GABABR on the peripheral endings of colonic afferents reduces nociceptive signalling. The enhanced antinociceptive actions of Vc1.1 during CVH suggest it is a novel candidate for the treatment for CVP.


Asunto(s)
Colon/fisiología , Conotoxinas/farmacología , Ganglios Espinales/fisiología , Neuronas Aferentes/fisiología , Nocicepción/efectos de los fármacos , Receptores de GABA-B/análisis , Receptores de GABA-B/genética , Animales , Baclofeno/farmacología , Canales de Calcio Tipo N/análisis , Canales de Calcio Tipo N/genética , Canales de Calcio Tipo N/metabolismo , Canales de Calcio Tipo R/análisis , Canales de Calcio Tipo R/genética , Canales de Calcio Tipo R/metabolismo , Proteínas de Transporte de Catión/análisis , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Células Cultivadas , Dolor Crónico/prevención & control , Modelos Animales de Enfermedad , Electrofisiología , Femenino , Agonistas de Receptores GABA-B/farmacología , Antagonistas de Receptores de GABA-B/farmacología , Ganglios Espinales/química , Ganglios Espinales/efectos de los fármacos , Expresión Génica , Humanos , Masculino , Ratones , Neuronas Aferentes/química , Neuronas Aferentes/efectos de los fármacos , Receptores de GABA-B/metabolismo , Regulación hacia Arriba , Dolor Visceral/prevención & control , Adulto Joven
15.
Gut ; 66(2): 258-269, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-26565000

RESUMEN

OBJECTIVE: The gut-brain axis is considered as a major regulatory checkpoint in the control of glucose homeostasis. The detection of nutrients and/or hormones in the duodenum informs the hypothalamus of the host's nutritional state. This process may occur via hypothalamic neurons modulating central release of nitric oxide (NO), which in turn controls glucose entry into tissues. The enteric nervous system (ENS) modulates intestinal contractions in response to various stimuli, but the importance of this interaction in the control of glucose homeostasis via the brain is unknown. We studied whether apelin, a bioactive peptide present in the gut, regulates ENS-evoked contractions, thereby identifying a new physiological partner in the control of glucose utilisation via the hypothalamus. DESIGN: We measured the effect of apelin on electrical and mechanical duodenal responses via telemetry probes and isotonic sensors in normal and obese/diabetic mice. Changes in hypothalamic NO release, in response to duodenal contraction modulated by apelin, were evaluated in real time with specific amperometric probes. Glucose utilisation in tissues was measured with orally administrated radiolabeled glucose. RESULTS: In normal and obese/diabetic mice, glucose utilisation is improved by the decrease of ENS/contraction activities in response to apelin, which generates an increase in hypothalamic NO release. As a consequence, glucose entry is significantly increased in the muscle. CONCLUSIONS: Here, we identify a novel mode of communication between the intestine and the hypothalamus that controls glucose utilisation. Moreover, our data identified oral apelin administration as a novel potential target to treat metabolic disorders.


Asunto(s)
Adipoquinas/farmacología , Sistema Nervioso Entérico/efectos de los fármacos , Glucosa/metabolismo , Hipotálamo/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Contracción Muscular/efectos de los fármacos , Animales , Apelina , Técnicas Biosensibles , Diabetes Mellitus/fisiopatología , Duodeno/efectos de los fármacos , Duodeno/metabolismo , Sistema Nervioso Entérico/fisiología , Motilidad Gastrointestinal/efectos de los fármacos , Homeostasis , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Liso/fisiología , Óxido Nítrico/metabolismo , Obesidad/fisiopatología , Telemetría
16.
Angew Chem Int Ed Engl ; 55(15): 4692-6, 2016 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-26948522

RESUMEN

α-Conotoxins are disulfide-rich peptides that target nicotinic acetylcholine receptors. Recently we identified several α-conotoxins that also modulate voltage-gated calcium channels by acting as G protein-coupled GABA(B) receptor (GABA(B)R) agonists. These α-conotoxins are promising drug leads for the treatment of chronic pain. To elucidate the diversity of α-conotoxins that act through this mechanism, we synthesized and characterized a set of peptides with homology to α-conotoxins known to inhibit high voltage-activated calcium channels via GABA(B)R activation. Remarkably, all disulfide isomers of the active α-conotoxins Pu1.2 and Pn1.2, and the previously studied Vc1.1 showed similar levels of biological activity. Structure determination by NMR spectroscopy helped us identify a simplified biologically active eight residue peptide motif containing a single disulfide bond that is an excellent lead molecule for developing a new generation of analgesic peptide drugs.


Asunto(s)
Secuencias de Aminoácidos , Bloqueadores de los Canales de Calcio/farmacología , Conotoxinas/química , Cisteína/análisis , Receptores de GABA-B/metabolismo , Secuencia de Aminoácidos , Animales , Conotoxinas/farmacología , Humanos , Receptores de GABA-B/química , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad , Xenopus
17.
Gastroenterology ; 145(6): 1334-46.e1-11, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23958540

RESUMEN

BACKGROUND & AIMS: Linaclotide is a minimally absorbed agonist of guanylate cyclase-C (GUCY2C or GC-C) that reduces symptoms associated with irritable bowel syndrome with constipation (IBS-C). Little is known about the mechanism by which linaclotide reduces abdominal pain in patients with IBS-C. METHODS: We determined the effects of linaclotide on colonic sensory afferents in healthy mice and those with chronic visceral hypersensitivity. We assessed pain transmission by measuring activation of dorsal horn neurons in the spinal cord in response to noxious colorectal distention. Levels of Gucy2c messenger RNA were measured in tissues from mice using quantitative reverse transcription polymerase chain reaction and in situ hybridization. We used human intestinal cell lines to measure release of cyclic guanosine-3',5'-monophosphate (cGMP) by linaclotide. We performed a post-hoc analysis of data from a phase III, double-blind, parallel-group study in which 805 patients with IBS-C were randomly assigned to groups given an oral placebo or 290 µg linaclotide once daily for 26 weeks. We quantified changes in IBS-C symptoms, including abdominal pain. RESULTS: In mice, linaclotide inhibited colonic nociceptors with greater efficacy during chronic visceral hypersensitivity. Intra-colonic administration of linaclotide reduced signaling of noxious colorectal distention to the spinal cord. The colonic mucosa, but not neurons, was found to express linaclotide's target, GC-C. The downstream effector of GC-C, cGMP, was released after administration of linaclotide and also inhibited nociceptors. The effects of linaclotide were lost in Gucy2c(-/-) mice and prevented by inhibiting cGMP transporters or removing the mucosa. During 26 weeks of linaclotide administration, a significantly greater percentage of patients (70%) had at least a 30% reduction in abdominal pain compared with patients given placebo (50%). CONCLUSIONS: We have identified an analgesic mechanism of linaclotide: it activates GC-C expressed on mucosal epithelial cells, resulting in the production and release of cGMP. This extracellular cGMP acts on and inhibits nociceptors, thereby reducing nociception. We also found that linaclotide reduces chronic abdominal pain in patients with IBS-C.


Asunto(s)
Dolor Abdominal/prevención & control , Colon/inervación , GMP Cíclico/fisiología , Guanilato Ciclasa/fisiología , Nociceptores/efectos de los fármacos , Péptidos/farmacología , Péptidos/uso terapéutico , Dolor Abdominal/inducido químicamente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Células CACO-2 , Línea Celular , Colon/efectos de los fármacos , Colon/patología , Modelos Animales de Enfermedad , Método Doble Ciego , Femenino , Humanos , Síndrome del Colon Irritable/inducido químicamente , Síndrome del Colon Irritable/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Péptidos Natriuréticos/farmacología , Nociceptores/fisiología , Receptores del Factor Natriurético Atrial/fisiología , Receptores de Enterotoxina , Receptores Acoplados a la Guanilato-Ciclasa/fisiología , Receptores de Péptidos/fisiología , Resultado del Tratamiento , Ácido Trinitrobencenosulfónico/efectos adversos
18.
Gut ; 62(10): 1456-65, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22767422

RESUMEN

OBJECTIVE: The gut is a major site of contact between immune and sensory systems and evidence suggests that patients with irritable bowel syndrome (IBS) have immune dysfunction. Here we show how this dysfunction differs between major IBS subgroups and how immunocytes communicate with sensory nerves. DESIGN: Peripheral blood mononuclear cell supernatants from 20 diarrhoea predominant IBS (D-IBS) patients, 15 constipation predominant IBS (C-IBS) patients and 36 healthy subjects were applied to mouse colonic sensory nerves and effects on mechanosensitivity assessed. Cytokine/chemokine concentration in the supernatants was assessed by proteomic analysis and correlated with abdominal symptoms, and expression of cytokine receptors evaluated in colonic dorsal root ganglia neurons. We then determined the effects of specific cytokines on colonic afferents. RESULTS: D-IBS supernatants caused mechanical hypersensitivity of mouse colonic afferent endings, which was reduced by infliximab. C-IBS supernatants did not, but occasionally elevated basal discharge. Supernatants of healthy subjects inhibited afferent mechanosensitivity via an opioidergic mechanism. Several cytokines were elevated in IBS supernatants, and levels correlated with pain frequency and intensity in patients. Visceral afferents expressed receptors for four cytokines: IL-1ß, IL-6, IL-10 and TNF-α. TNF-α most effectively caused mechanical hypersensitivity which was blocked by a transient receptor potential channel TRPA1 antagonist. IL-1ß elevated basal firing, and this was lost after tetrodotoxin blockade of sodium channels. CONCLUSIONS: Distinct patterns of immune dysfunction and interaction with sensory pathways occur in different patient groups and through different intracellular pathways. Our results indicate IBS patient subgroups would benefit from selective targeting of the immune system.


Asunto(s)
Síndrome del Colon Irritable/inmunología , Neuroinmunomodulación/fisiología , Adulto , Animales , Estudios de Casos y Controles , Células Cultivadas , Colon/inmunología , Colon/inervación , Estreñimiento/etiología , Estreñimiento/inmunología , Medios de Cultivo Condicionados/farmacología , Citocinas/biosíntesis , Diarrea/etiología , Diarrea/inmunología , Femenino , Ganglios Espinales/inmunología , Humanos , Síndrome del Colon Irritable/complicaciones , Síndrome del Colon Irritable/fisiopatología , Leucocitos Mononucleares/metabolismo , Masculino , Ratones , Persona de Mediana Edad , Neuroinmunomodulación/inmunología , Neuronas Aferentes/efectos de los fármacos , Neuronas Aferentes/fisiología , Dolor/etiología , Dolor/inmunología , Receptores de Citocinas/metabolismo , betaendorfina/metabolismo
19.
Pain ; 164(5): 1012-1026, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36279179

RESUMEN

ABSTRACT: The bladder wall is innervated by a complex network of afferent nerves that detect bladder stretch during filling. Sensory signals, generated in response to distension, are relayed to the spinal cord and brain to evoke physiological and painful sensations and regulate urine storage and voiding. Hyperexcitability of these sensory pathways is a key component in the development of chronic bladder hypersensitivity disorders including interstitial cystitis/bladder pain syndrome and overactive bladder syndrome. Despite this, the full array of ion channels that regulate bladder afferent responses to mechanical stimuli have yet to be determined. Here, we investigated the role of low-voltage-activated T-type calcium (Ca V 3) channels in regulating bladder afferent responses to distension. Using single-cell reverse-transcription polymerase chain reaction and immunofluorescence, we revealed ubiquitous expression of Ca V 3.2, but not Ca V 3.1 or Ca V 3.3, in individual bladder-innervating dorsal root ganglia neurons. Pharmacological inhibition of Ca V 3.2 with TTA-A2 and ABT-639, selective blockers of T-type calcium channels, dose-dependently attenuated ex-vivo bladder afferent responses to distension in the absence of changes to muscle compliance. Further evaluation revealed that Ca V 3.2 blockers significantly inhibited both low- and high-threshold afferents, decreasing peak responses to distension, and delayed activation thresholds, thereby attenuating bladder afferent responses to both physiological and noxious distension. Nocifensive visceromotor responses to noxious bladder distension in vivo were also significantly reduced by inhibition of Ca V 3 with TTA-A2. Together, these data provide evidence of a major role for Ca V 3.2 in regulating bladder afferent responses to bladder distension and nociceptive signalling to the spinal cord.


Asunto(s)
Canales de Calcio Tipo T , Cistitis Intersticial , Humanos , Vejiga Urinaria/inervación , Neuronas Aferentes/fisiología , Canales de Calcio Tipo T/metabolismo , Vías Aferentes/fisiología , Cistitis Intersticial/metabolismo , Ganglios Espinales/metabolismo
20.
Pain ; 163(8): 1622-1635, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35050959

RESUMEN

ABSTRACT: Endometriosis is a chronic and debilitating condition, commonly characterised by chronic pelvic pain (CPP) and infertility. Chronic pelvic pain can be experienced across multiple pelvic organs, with comorbidities commonly effecting the bowel, bladder, and vagina. Despite research efforts into endometriosis pathophysiology, little is known about how endometriosis induces CPP, and as such, therapeutic interventions are lacking. The aim of this study was to characterise a syngeneic mouse model of endometriosis that mimics naturally occurring retrograde menstruation, thought to precede endometriosis development in patients, and determine whether these mice exhibit signs of CPP and altered behaviour. We characterised the development of endometriosis over 10 weeks following uterine tissue inoculation, measured in vivo and ex vivo hypersensitivity to mechanical stimuli across multiple visceral organs, and assessed alterations in animal spontaneous behaviour. We confirmed that inoculated uterine horn tissue formed into endometriosis lesions throughout the peritoneal cavity, with significant growth by 8 to 10 weeks post inoculation. Additionally, we found that mice with fully developed endometriosis displayed hypersensitivity evoked by (1) vaginal distension, (2) colorectal distension, (3) bladder distension, and (4) cutaneous thermal stimulation, compared to their sham counterparts. Moreover, endometriosis mice displayed alterations in spontaneous behaviour indicative of (5) altered bladder function and (6) anxiety. This model creates a foundation for mechanistical studies into the diffuse CPP associated with endometriosis and the development of targeted therapeutic interventions to improve the quality of life of women with endometriosis.


Asunto(s)
Dolor Crónico , Endometriosis , Animales , Dolor Crónico/complicaciones , Modelos Animales de Enfermedad , Endometriosis/complicaciones , Femenino , Humanos , Ratones , Dolor Pélvico/diagnóstico , Calidad de Vida
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA