Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros











Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(18): e2123020119, 2022 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-35446689

RESUMEN

The peristaltic reflex is a fundamental behavior of the gastrointestinal (GI) tract in which mucosal stimulation activates propulsive contractions. The reflex occurs by stimulation of intrinsic primary afferent neurons with cell bodies in the myenteric plexus and projections to the lamina propria, distribution of information by interneurons, and activation of muscle motor neurons. The current concept is that excitatory cholinergic motor neurons are activated proximal to and inhibitory neurons are activated distal to the stimulus site. We found that atropine reduced, but did not block, colonic migrating motor complexes (CMMCs) in mouse, monkey, and human colons, suggesting a mechanism other than one activated by cholinergic neurons is involved in the generation/propagation of CMMCs. CMMCs were activated after a period of nerve stimulation in colons of each species, suggesting that the propulsive contractions of CMMCs may be due to the poststimulus excitation that follows inhibitory neural responses. Blocking nitrergic neurotransmission inhibited poststimulus excitation in muscle strips and blocked CMMCs in intact colons. Our data demonstrate that poststimulus excitation is due to increased Ca2+ transients in colonic interstitial cells of Cajal (ICC) following cessation of nitrergic, cyclic guanosine monophosphate (cGMP)-dependent inhibitory responses. The increase in Ca2+ transients after nitrergic responses activates a Ca2+-activated Cl− conductance, encoded by Ano1, in ICC. Antagonists of ANO1 channels inhibit poststimulus depolarizations in colonic muscles and CMMCs in intact colons. The poststimulus excitatory responses in ICC are linked to cGMP-inhibited cyclic adenosine monophosphate (cAMP) phosphodiesterase 3a and cAMP-dependent effects. These data suggest alternative mechanisms for generation and propagation of CMMCs in the colon.


Asunto(s)
Células Intersticiales de Cajal , Colon/fisiología , Motilidad Gastrointestinal/fisiología , Miocitos del Músculo Liso , Peristaltismo
2.
Sci Rep ; 12(1): 5071, 2022 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-35332235

RESUMEN

Cyclophosphamide (CYP)-induced cystitis is a rodent model that shares many features common to the cystitis occurring in patients, including detrusor overactivity (DO). Platelet-derived growth factor receptor alpha positive (PDGFRα+) cells have been proposed to regulate muscle excitability in murine bladders during filling. PDGFRα+ cells express small conductance Ca2+-activated K+ channels (predominantly SK3) that provide stabilization of membrane potential during filling. We hypothesized that down-regulation of the regulatory functions of PDGFRα+ cells and/or loss of PDGFRα+ cells generates the DO in CYP-treated mice. After CYP treatment, transcripts of Pdgfrα and Kcnn3 and PDGFRα and SK3 protein were reduced in detrusor muscle extracts. The distribution of PDGFRα+ cells was also reduced. Inflammatory markers were increased in CYP-treated detrusor muscles. An SK channel agonist, CyPPA, increased outward current and hyperpolarization in PDGFRα+ cells. This response was significantly depressed in PDGFRα+ cells from CYP-treated bladders. Contractile experiments and ex vivo cystometry showed increased spontaneous contractions and transient contractions, respectively in CYP-treated bladders with a reduction of apamin sensitivity, that could be attributable to the reduction in the SK conductance expressed by PDGFRα+ cells. In summary, PDGFRα+ cells were reduced and the SK3 conductance was downregulated in CYP-treated bladders. These changes are consistent with the development of DO after CYP treatment.


Asunto(s)
Cistitis , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas , Animales , Apamina , Ciclofosfamida/efectos adversos , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo
3.
FASEB J ; 34(4): 5563-5577, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32086857

RESUMEN

Transcriptome data revealed α1 adrenoceptors (ARs) expression in platelet-derived growth factor receptor α+ cells (PDGFRα+ cells) in murine colonic musculature. The role of PDGFRα+ cells in sympathetic neural regulation of murine colonic motility was investigated. Norepinephrine (NE), via α1A ARs, activated a small conductance Ca2+ -activated K+ (SK) conductance, evoked outward currents and hyperpolarized PDGFRα+ cells (the α1A AR-SK channel signal pathway). α1 AR agonists increased intracellular Ca2+ transients in PDGFRα+ cells and inhibited spontaneous phasic contractions (SPCs) of colonic muscle through activation of a SK conductance. Sympathetic nerve stimulation inhibited both contractions of distal colon and propulsive contractions represented by the colonic migrating motor complexes (CMMCs) via the α1A AR-SK channel signal pathway. Postsynaptic signaling through α1A ARs in PDGFRα+ cells is a novel mechanism that conveys part of stress responses in the colon. PDGFRα+ cells appear to be a primary effector of sympathetic neural regulation of murine colonic motility.


Asunto(s)
Colon/fisiología , Músculo Liso/fisiología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptores Adrenérgicos alfa 1/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Sistema Nervioso Simpático/fisiología , Potenciales Sinápticos , Adenosina Trifosfato , Animales , Calcio/metabolismo , Colon/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Liso/citología , Transducción de Señal , Sistema Nervioso Simpático/citología
4.
Transl Oncol ; 13(2): 233-240, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31869747

RESUMEN

BACKGROUND: Alpha-type platelet-derived growth factor receptor (PDGFRα) is a cell surface tyrosine kinase receptor for members of the platelet-derived growth factor family. PDGFRα plays an important role in the regulation of several biological processes and contributes to the pathophysiology of a broad range of human cancers, including glioma. Here, we hypothesize that the genes directly or indirectly influenced by PDGFRα might be useful for prognosis in glioma. METHODS: By comparing the genome-wide gene expression pattern between PDGFRα+ and PDGFRα- cells from human oligodendrocyte progenitor, we defined the genes potentially influenced by PDGFRα. RESULTS: The PDGFRα-influenced genes are strongly associated with cancer-related pathways. We subsequently developed a prognostic gene signature derived from the PDGFRα-influenced genes. This gene signature is able to predict clinical outcome of glioma. This signature is also independent of traditional prognostic factors of glioma. Resampling tests indicate that the prognostic power of this gene signature outperforms random gene sets selected from human genome. More importantly, this signature is superior to the random gene signatures selected from glioma related genes. CONCLUSIONS: Despite the absence of clear elucidation of molecular mechanisms, this study suggests the vital role of PDGFRα in carcinogenesis. Furthermore, the PDGFRα-based gene signature provides a promising prognostic tool for glioma and validates PDGFRα as a novel and effective therapeutic target in human cancers.

5.
Sci Rep ; 9(1): 6446, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-31015490

RESUMEN

Recent parallel studies clearly indicated that Merkel cells and the mechanosensitive piezo2 ion channel play critical roles in the light-touch somatosensation. Moreover, piezo2 was suggested to be a light-touch sensing ion channel without a role in pain sensing in mammals. However, biophysical characteristics of piezo2, such as single channel conductance and sensitivities to various mechanical stimuli, are unclear, hampering a precise understanding of its role in touch sensation. Here, we describe the biophysical properties of piezo2 in human Merkel cell carcinoma (MCC)-13 cells; piezo2 is a low-threshold, positive pressure-specific, curvature-sensitive, mechanically activated cation channel with a single channel conductance of ~28.6 pS. Application of step indentations under the whole-cell mode of the patch-clamp technique, and positive pressures ≥5 mmHg under the cell-attached mode, activated piezo2 currents in MCC-13 and human embryonic kidney 293 T cells where piezo2 was overexpressed. By contrast, application of a negative pressure failed to activate piezo2 in these cells, whereas both positive and negative pressure activated piezo1 in a similar manner. Our results are the first to demonstrate single channel recordings of piezo2. We anticipate that our findings will be a starting point for a more sophisticated understanding of piezo2 roles in light-touch sensation.


Asunto(s)
Canales Iónicos/metabolismo , Presión , Tacto , Línea Celular Tumoral , Células HEK293 , Humanos , Mecanotransducción Celular
6.
J Physiol ; 597(6): 1467-1485, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30289177

RESUMEN

KEY POINTS: Studies of urothelial cells, bladder sheets or lumens of filled bladders have suggested that mediators released from urothelium into suburothelium (SubU)/lamina propria (LP) activate mechanisms controlling detrusor excitability. None of these approaches, however, has enabled direct assessment of availability of mediators at SubU/LP during filling. We developed an ex vivo mouse bladder preparation with intact urothelium and SubU/LP but no detrusor, which allows direct access to the SubU/LP surface of urothelium during filling. Pressure-volume measurements during filling demonstrated that bladder compliance is governed primarily by the urothelium. Measurements of purine mediators in this preparation demonstrated asymmetrical availability of purines in lumen and SubU/LP, suggesting that interpretations based solely on intraluminal measurements of mediators may be inaccurate. The preparations are suitable for assessments of release, degradation and transport of mediators in SubU/LP during bladder filling, and are superior to experimental approaches previously used for urothelium research. ABSTRACT: The purpose of this study was to develop a decentralized (ex vivo) detrusor smooth muscle (DSM)-denuded mouse bladder preparation, a novel model that enables studies on availability of urothelium-derived mediators at the luminal and anti-luminal aspects of the urothelium during filling. Urinary bladders were excised from C57BL6/J mice and the DSM was removed by fine-scissor dissection without touching the mucosa. Morphology and cell composition of the preparation wall, pressure-volume relationships during filling, and fluorescent dye permeability of control, protamine sulfate- and lipopolysaccharide-treated denuded bladders were characterized. The preparation wall contained intact urothelium and suburothelium (SubU)/lamina propria (LP) and lacked the DSM and the serosa. The utility of the model for physiological research was validated by measuring release, metabolism and transport of purine mediators at SubU/LP and in bladder lumen during filling. We determined asymmetrical availability of purines (e.g. ATP, ADP, AMP and adenosine) in lumen and at SubU/LP during filling, suggesting differential mechanisms of release, degradation and bilateral transurothelial transport of purines during filling. Some observations were validated in DSM-denuded bladder of the cynomolgus monkey (Macaca fascicularis). The novel model was superior to current models utilized to study properties of the urothelium (e.g. cultured urothelial cells, bladder mucosa sheets mounted in Ussing chambers or isolated bladder strips in organ baths) in that it enabled direct access to the vicinity of SubU/LP during authentic bladder filling. The model is particularly suitable for understanding local mechanisms of urothelium-DSM connectivity and for broad understanding of the role of urothelium in regulating continence and voiding.


Asunto(s)
Músculo Liso/fisiología , Vejiga Urinaria/fisiología , Urotelio/fisiología , Animales , Femenino , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Músculo Liso/citología , Músculo Liso/metabolismo , Técnicas de Cultivo de Órganos/métodos , Purinas/metabolismo , Vejiga Urinaria/citología , Vejiga Urinaria/metabolismo , Urotelio/citología , Urotelio/metabolismo
7.
Sci Signal ; 11(534)2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29895614

RESUMEN

Electrical pacemaker activity generates phasic contractions and motility patterns such as segmentation and peristalsis in the gastrointestinal tract. Pacemaker currents are generated in interstitial cells of Cajal (ICC), which release Ca2+ from intracellular stores that stimulates Ca2+-activated Cl- channels (CaCCs) in the plasma membrane. Thus, Ca2+ stores must be maintained to sustain pacemaker activity. Store-operated Ca2+ entry (SOCE) facilitates the refilling of Ca2+ stores by a mechanism dependent upon interactions between STIM and Orai proteins. We investigated the role of SOCE in ICC pacemaker activity. Reintroduction of extracellular Ca2+ in store-depleted ICC resulted in CaCC activation. Blocking CaCCs revealed an inwardly rectifying current with properties of a Ca2+ release-activated current (ICRAC). An inhibitory peptide that interfered with the STIM-Orai interaction blocked ICRAC in HEK 293 cells expressing STIM1 and Orai1 and blocked spontaneous transient inward currents (STICs) and slow wave currents in ICC. STICs, which are fundamental pacemaker events in ICC, were blocked by an Orai antagonist. Imaging of Ca2+ transients linked to pacemaker activity in ICC in intact muscles showed that the Orai antagonist blocked Ca2+ transients in ICC. These data suggest that Ca2+ recovery through STIM-Orai interactions is necessary to maintain ICC pacemaker activity.


Asunto(s)
Relojes Biológicos , Canales de Calcio/metabolismo , Tracto Gastrointestinal/metabolismo , Células Intersticiales de Cajal/metabolismo , Moléculas de Interacción Estromal/metabolismo , Animales , Calcio/metabolismo , Señalización del Calcio , Células Cultivadas , Canales de Cloruro/metabolismo , Tracto Gastrointestinal/citología , Células HEK293 , Humanos , Células Intersticiales de Cajal/citología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo , Moléculas de Interacción Estromal/genética
8.
Am J Physiol Renal Physiol ; 311(4): F708-F716, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27465992

RESUMEN

During urinary bladder filling the bladder urothelium releases chemical mediators that in turn transmit information to the nervous and muscular systems to regulate sensory sensation and detrusor muscle activity. Defects in release of urothelial mediators may cause bladder dysfunctions that are characterized with aberrant bladder sensation during bladder filling. Previous studies have demonstrated release of ATP from the bladder urothelium during bladder filling, and ATP remains the most studied purine mediator that is released from the urothelium. However, the micturition cycle is likely regulated by multiple purine mediators, since various purine receptors are found present in many cell types in the bladder wall, including urothelial cells, afferent nerves, interstitial cells in lamina propria, and detrusor smooth muscle cells. Information about the release of other biologically active purines during bladder filling is still lacking. Decentralized bladders from C57BL/6 mice and Cynomolgus monkeys (Macaca fascicularis) were filled with physiological solution at different rates. Intraluminal fluid was analyzed by high-performance liquid chromatography with fluorescence detection for simultaneous evaluation of ATP, ADP, AMP, adenosine, nicotinamide adenine dinucleotide (NAD+), ADP-ribose, and cADP-ribose content. We also measured ex vivo bladder filling pressures and performed cystometry in conscious unrestrained mice at different filling rates. ATP, ADP, AMP, NAD+, ADPR, cADPR, and adenosine were detected released intravesically at different ratios during bladder filling. Purine release increased with increased volumes and rates of filling. Our results support the concept that multiple urothelium-derived purines likely contribute to the complex regulation of bladder sensation during bladder filling.


Asunto(s)
Músculo Liso/fisiología , Purinas/metabolismo , Receptores Purinérgicos/metabolismo , Vejiga Urinaria/fisiología , Micción/fisiología , Urotelio/metabolismo , Animales , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Liso/metabolismo , Sensación/fisiología , Vejiga Urinaria/metabolismo
9.
Am J Physiol Renal Physiol ; 309(6): F569-74, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26202222

RESUMEN

Purines induce transient contraction and prolonged relaxation of detrusor muscles. Transient contraction is likely due to activation of inward currents in smooth muscle cells, and prolonged relaxation may be due to activation of small-conductance Ca(2+)-activated K(+) (SK) channels via P2Y1 receptors expressed by detrusor PDGF receptor (PDGFR)α(+) cells. We investigated whether other subtypes of P2Y receptors are involved in the activation of SK channels in PDGFRα(+) cells of detrusor muscles. Quantitative analysis of transcripts revealed that P2ry2, P2ry4, and P2ry14 are expressed in PDGFRα(+) cells of P2ry1-deficient/enhanced green fluorescent protein (P2ry1(-/-)/eGFP) mice at similar levels as in wild-type mice. UTP, a P2Y2/P2Y4 agonist, activated large outward currents in detrusor PDGFRα(+) cells. SK channel blockers and an inhibitor of phospholipase C completely abolished currents activated by UTP. In contrast, UTP activated nonselective cation currents in smooth muscle cells. Under current-clamp (current = 0), UTP induced significant hyperpolarization of PDGFRα(+) cells. MRS2500, a selective P2Y1 antagonist, did not affect UTP-activated outward currents in PDGFRα(+) cells from wild-type mice, and activation of outward currents by UTP was retained in P2ry1(-/-)/eGFP mice. As a negative control, we tested the effect of MRS2693, a selective P2Y6 agonist. This compound did not activate outward currents in PDGFRα(+) cells, and currents activated by UTP were unaffected by MRS2578, a selective P2Y6 antagonist. The nonselective P2Y receptor blocker suramin inhibited UTP-activated outward currents in PDGFRα(+) cells. Our data demonstrate that P2Y2 and/or P2Y4 receptors function, in addition to P2Y1 receptors, in activating SK currents in PDGFRα(+) cells and possibly in mediating purinergic relaxation responses in detrusor muscles.


Asunto(s)
Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/agonistas , Uridina Trifosfato/farmacología , Vejiga Urinaria/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Ratones , Músculo Liso/metabolismo , Miocitos del Músculo Liso/metabolismo , Técnicas de Placa-Clamp , Receptores Purinérgicos P2/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Receptores Purinérgicos P2Y2/metabolismo , Transducción de Señal/efectos de los fármacos , Vejiga Urinaria/efectos de los fármacos
10.
Am J Physiol Cell Physiol ; 308(8): C608-20, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25631870

RESUMEN

Interstitial cells of Cajal (ICC) provide pacemaker activity in gastrointestinal muscles that underlies segmental and peristaltic contractions. ICC generate electrical slow waves that are due to large-amplitude inward currents resulting from anoctamin 1 (ANO1) channels, which are Ca(2+)-activated Cl(-) channels. We investigated the hypothesis that the Ca(2+) responsible for the stochastic activation of ANO1 channels during spontaneous transient inward currents (STICs) and synchronized activation of ANO1 channels during slow wave currents comes from intracellular Ca(2+) stores. ICC, obtained from the small intestine of Kit(+/copGFP) mice, were studied under voltage and current clamp to determine the effects of blocking Ca(2+) uptake into stores and release of Ca(2+) via inositol 1,4,5-trisphosphate (IP3)-dependent and ryanodine-sensitive channels. Cyclocpiazonic acid, thapsigargin, 2-APB, and xestospongin C inhibited STICs and slow wave currents. Ryanodine and tetracaine also inhibited STICs and slow wave currents. Store-active compounds had no direct effects on ANO1 channels expressed in human embryonic kidney-293 cells. Under current clamp, store-active drugs caused significant depolarization of ICC and reduced spontaneous transient depolarizations (STDs). After block of ryanodine receptors with ryanodine and tetracaine, repolarization did not restore STDs. ANO1 expressed in ICC has limited access to cytoplasmic Ca(2+) concentration, suggesting that pacemaker activity depends on Ca(2+) dynamics in restricted microdomains. Our data from studies of isolated ICC differ somewhat from studies on intact muscles and suggest that release of Ca(2+) from both IP3 and ryanodine receptors is important in generating pacemaker activity in ICC.


Asunto(s)
Calcio/metabolismo , Canales de Cloruro/metabolismo , Retículo Endoplásmico/metabolismo , Células Intersticiales de Cajal/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Animales , Anoctamina-1 , Bloqueadores de los Canales de Calcio/farmacología , ATPasas Transportadoras de Calcio/antagonistas & inhibidores , Células Cultivadas , Canales de Cloruro/biosíntesis , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Células HEK293 , Humanos , Indoles/farmacología , Inositol 1,4,5-Trifosfato/química , Intestino Delgado/citología , Compuestos Macrocíclicos/farmacología , Potenciales de la Membrana/efectos de los fármacos , Ratones , Contracción Muscular/fisiología , Miocitos del Músculo Liso/metabolismo , Oxazoles/farmacología , Técnicas de Placa-Clamp , Rianodina/farmacología , Tapsigargina/farmacología
11.
Am J Physiol Cell Physiol ; 307(6): C561-70, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25055825

RESUMEN

Enteric inhibitory neurotransmission is an important feature of the neural regulation of gastrointestinal motility. Purinergic neurotransmission, via P2Y1 receptors, mediates one phase of inhibitory neural control. For decades, ATP has been assumed to be the purinergic neurotransmitter and smooth muscle cells (SMCs) have been considered the primary targets for inhibitory neurotransmission. Recent experiments have cast doubt on both of these assumptions and suggested that another cell type, platelet-derived growth factor receptor-α-positive (PDGFRα(+)) cells, is the target for purinergic neurotransmission. We compared responses of PDGFRα(+) cells and SMCs to several purine compounds to determine if these cells responded in a manner consistent with enteric inhibitory neurotransmission. ATP hyperpolarized PDGFRα(+) cells but depolarized SMCs. Only part of the ATP response in PDGFRα(+) cells was blocked by MRS 2500, a P2Y1 antagonist. ADP, MRS 2365, ß-NAD, and adenosine 5-diphosphate-ribose, P2Y1 agonists, hyperpolarized PDGFRα(+) cells, and these responses were blocked by MRS 2500. Adenosine 5-diphosphate-ribose was more potent in eliciting hyperpolarization responses than ß-NAD. P2Y1 agonists failed to elicit responses in SMCs. Small hyperpolarization responses were elicited in SMCs by a small-conductance Ca(2+)-activated K(+) channel agonist, cyclohexyl-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-pyrimidin-4-yl]-amine, consistent with the low expression and current density of small-conductance Ca(2+)-activated K(+) channels in these cells. Large-amplitude hyperpolarization responses, elicited in PDGFRα(+) cells, but not SMCs, by P2Y1 agonists are consistent with the generation of inhibitory junction potentials in intact muscles in response to purinergic neurotransmission. The responses of PDGFRα(+) cells and SMCs to purines suggest that SMCs are unlikely targets for purinergic neurotransmission in colonic muscles.


Asunto(s)
Adenosina Difosfato/metabolismo , Adenosina Trifosfato/metabolismo , Colon/metabolismo , Músculo Liso/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Colon/citología , Colon/efectos de los fármacos , Colon/inervación , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Potenciales de la Membrana , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Liso/citología , Músculo Liso/efectos de los fármacos , Músculo Liso/inervación , Miocitos del Músculo Liso/efectos de los fármacos , Inhibición Neural , Técnicas de Placa-Clamp , Agonistas del Receptor Purinérgico P2Y/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptores Purinérgicos P2Y1/efectos de los fármacos , Receptores Purinérgicos P2Y1/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/agonistas , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo
12.
J Physiol ; 592(6): 1283-93, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24396055

RESUMEN

Purines induce transient contraction and prolonged relaxation of detrusor muscles. Transient contraction could be due to activation of inward currents in smooth muscle cells, but the mechanism of purinergic relaxation has not been determined. We recently reported a new class of interstitial cells in detrusor muscles and showed that these cells could be identified with antibodies against platelet-derived growth factor receptor-α (PDGFRα(+) cells). The current density of small conductance Ca(2+)-activated K(+) (SK) channels in these cells is far higher (∼100 times) than in smooth muscle cells. Thus, we examined purinergic receptor (P2Y) mediated SK channel activation as a mechanism for purinergic relaxation. P2Y receptors (mainly P2ry1 gene) were highly expressed in PDGFRα(+) cells. Under voltage clamp conditions, ATP activated large outward currents in PDGFRα(+) cells that were inhibited by blockers of SK channels. ATP also induced significant hyperpolarization under current clamp conditions. A P2Y1 agonist, MRS2365, mimicked the effects of ATP, and a P2Y1 antagonist, MRS2500, inhibited ATP-activated SK currents. Responses to ATP were largely abolished in PDGFRα(+) cells of P2ry1(-/-) mice, and no response was elicited by MRS2365 in these cells. A P2X receptor agonist had no effect on PDGFRα(+) cells but, like ATP, activated transient inward currents in smooth muscle cells (SMCs). A P2Y1 antagonist decreased nerve-evoked relaxation. These data suggest that purines activate SK currents via mainly P2Y1 receptors in PDGFRα(+) cells. Our findings provide an explanation for purinergic relaxation in detrusor muscles and show that there are no discrete inhibitory nerve fibres. A dual receptive field for purines provides the basis for inhibitory neural regulation of excitability.


Asunto(s)
Músculo Liso/fisiología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/fisiología , Receptores Purinérgicos P2Y1/fisiología , Vejiga Urinaria/fisiología , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/farmacología , Adenosina Trifosfato/metabolismo , Adenosina Trifosfato/farmacología , Animales , Nucleótidos de Desoxiadenina/farmacología , Estimulación Eléctrica , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Relajación Muscular/efectos de los fármacos , Relajación Muscular/fisiología , Músculo Liso/efectos de los fármacos , Agonistas del Receptor Purinérgico P2Y/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y1/deficiencia , Receptores Purinérgicos P2Y1/genética , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Vejiga Urinaria/citología , Vejiga Urinaria/efectos de los fármacos
13.
Am J Physiol Heart Circ Physiol ; 305(4): H484-93, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23792675

RESUMEN

In endothelial cells (ECs), Ca²âº-activated K⁺ channels KCa2.3 and KCa3.1 play a crucial role in the regulation of arterial tone via producing NO and endothelium-derived hyperpolarizing factors. Since a rise in intracellular Ca²âº levels and activation of p300 histone acetyltransferase are early EC responses to laminar shear stress (LS) for the transcriptional activation of genes, we examined the role of Ca²âº/calmodulin-dependent kinase kinase (CaMKK), the most upstream element of a Ca²âº/calmodulin-kinase cascade, and p300 in LS-dependent regulation of KCa2.3 and KCa3.1 in ECs. Exposure to LS (15 dyn/cm²) for 24 h markedly increased KCa2.3 and KCa3.1 mRNA expression in cultured human coronary artery ECs (3.2 ± 0.4 and 45 ± 10 fold increase, respectively; P < 0.05 vs. static condition; n = 8-30), whereas oscillatory shear (OS; ± 5 dyn/cm² × 1 Hz) moderately increased KCa3.1 but did not affect KCa2.3. Expression of KCa2.1 and KCa2.2 was suppressed under both LS and OS conditions, whereas KCa1.1 was slightly elevated in LS and unchanged in OS. Inhibition of CaMKK attenuated LS-induced increases in the expression and channel activity of KCa2.3 and KCa3.1, and in phosphorylation of Akt (Ser473) and p300 (Ser1834). Inhibition of Akt abolished the upregulation of these channels by diminishing p300 phosphorylation. Consistently, disruption of the interaction of p300 with transcription factors eliminated the induction of these channels. Thus a CaMKK/Akt/p300 cascade plays an important role in LS-dependent induction of KCa2.3 and KCa3.1 expression, thereby regulating EC function and adaptation to hemodynamic changes.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Proteína p300 Asociada a E1A/metabolismo , Células Endoteliales/enzimología , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Mecanotransducción Celular , Proteínas Proto-Oncogénicas c-akt/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Adaptación Fisiológica , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/antagonistas & inhibidores , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Activación Enzimática , Hemodinámica , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Mecanotransducción Celular/efectos de los fármacos , Potenciales de la Membrana , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , ARN Mensajero/metabolismo , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética , Estrés Mecánico , Factores de Tiempo , Regulación hacia Arriba
14.
Nat Rev Gastroenterol Hepatol ; 9(11): 633-45, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22965426

RESUMEN

Gastrointestinal motility results from coordinated contractions of the tunica muscularis, the muscular layers of the alimentary canal. Throughout most of the gastrointestinal tract, smooth muscles are organized into two layers of circularly or longitudinally oriented muscle bundles. Smooth muscle cells form electrical and mechanical junctions between cells that facilitate coordination of contractions. Excitation-contraction coupling occurs by Ca(2+) entry via ion channels in the plasma membrane, leading to a rise in intracellular Ca(2+). Ca(2+) binding to calmodulin activates myosin light chain kinase; subsequent phosphorylation of myosin initiates cross-bridge cycling. Myosin phosphatase dephosphorylates myosin to relax muscles, and a process known as Ca(2+) sensitization regulates the activity of the phosphatase. Gastrointestinal smooth muscles are 'autonomous' and generate spontaneous electrical activity (slow waves) that does not depend upon input from nerves. Intrinsic pacemaker activity comes from interstitial cells of Cajal, which are electrically coupled to smooth muscle cells. Patterns of contractile activity in gastrointestinal muscles are determined by inputs from enteric motor neurons that innervate smooth muscle cells and interstitial cells. Here we provide an overview of the cells and mechanisms that generate smooth muscle contractile behaviour and gastrointestinal motility.


Asunto(s)
Motilidad Gastrointestinal/fisiología , Contracción Muscular/fisiología , Músculo Liso/fisiología , Calcio/fisiología , Humanos , Células Intersticiales de Cajal/fisiología , Miosinas/fisiología
15.
Exp Toxicol Pathol ; 64(5): 521-6, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21185163

RESUMEN

Recent studies of diabetic animal models suggest an important role of ICC in the pathogenesis of gastropathy. The aim of this study was to characterize the ultrastructural features of ICC and record the electrical properties in the stomach of patients with type 2 DM. Gastric specimens were obtained from 13 diabetic patients and 6 control subjects with gastric cancer that underwent gastrectomy. All specimens were taken from disease-free areas. The samples were processed for both electron microscopic and electrophysiologic examination. The characteristic ultrastructural changes of the ICC were observed in both the nucleus and cytoplasm in patients with type 2 DM. Wrinkling of the nuclear envelope and changes in the cytoplasm such as dilatation of the endoplasmic reticulum, an increase of autophagic vacuoles, were more frequently observed in the diabetic patients. Apoptosis characterized by nuclear karyorrhexis or pyknosis was observed only in the diabetic patients. Slow waves were recorded in the circular muscle of stomach. In diabetic patients, the mean resting membrane potential was higher and amplitude was lower than controls. These changes of electrical activities of slow waves were accompanied with ultrastructural changes of ICC, particularly the characteristic nuclear changes. In human diabetic patients, the characteristic ultrastructural changes of ICC such as preapoptosis, accompanied with electrical dysrhythmia of slow waves, were observed. These results show several evidence converging to support that degeneration of the ICC may be associated with the pathogenesis of diabetic gastropathy.


Asunto(s)
Diabetes Mellitus Tipo 2/complicaciones , Células Intersticiales de Cajal/ultraestructura , Gastropatías/etiología , Gastropatías/patología , Gastropatías/fisiopatología , Anciano , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/fisiopatología , Electrofisiología , Femenino , Humanos , Masculino , Potenciales de la Membrana , Microscopía Electrónica de Transmisión , Persona de Mediana Edad
16.
J Physiol ; 589(Pt 24): 6105-18, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22005683

RESUMEN

Extracellular electrical recording and studies using animal models have helped establish important concepts of human gastric physiology. Accepted standards include electrical quiescence in the fundus, 3 cycles per minute (cpm) pacemaker activity in corpus and antrum, and a proximal-to-distal slow wave frequency gradient. We investigated slow wave pacemaker activity, contractions and distribution of interstitial cells of Cajal (ICC) in human gastric muscles. Muscles were obtained from patients undergoing gastric resection for cancer, and the anatomical locations of each specimen were mapped by the operating surgeon to 16 standardized regions of the stomach. Electrical slow waves were recorded with intracellular microelectrodes and contractions were recorded by isometric force techniques. Slow waves were routinely recorded from gastric fundus muscles. These events had similar waveforms as slow waves in more distal regions and were coupled to phasic contractions. Gastric slow wave frequency was significantly greater than 3 cpm in all regions of the stomach. Antral slow wave frequency often exceeded the highest frequency of pacemaker activity in the corpus. Chronotropic mechanisms such as muscarinic and prostaglandin receptor binding, stretch, extracelluar Ca(2+) and temperature were unable to explain the observed slow wave frequency that exceeded accepted normal levels. Muscles from all regions through the thickness of the muscularis demonstrated intrinsic pacemaker activity, and this corresponded with the widespread distribution in ICC we mapped throughout the tunica muscularis. Our findings suggest that extracellular electrical recording has underestimated human slow wave frequency and mechanisms of human gastric function may differ from standard laboratory animal models.


Asunto(s)
Relojes Biológicos/fisiología , Músculo Liso/fisiología , Estómago/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Calcio/fisiología , Femenino , Humanos , Técnicas In Vitro , Células Intersticiales de Cajal/fisiología , Masculino , Persona de Mediana Edad , Contracción Muscular/fisiología
17.
Gastroenterology ; 140(2): 608-617.e6, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20875415

RESUMEN

BACKGROUND & AIMS: An important component of enteric inhibitory neurotransmission is mediated by a purine neurotransmitter, such as adenosine 5'-triphosphate (ATP), binding to P2Y1 receptors and activating small conductance K(+) channels. In murine colon ß-nicotinamide adenine dinucleotide (ß-NAD) is released with ATP and mimics the pharmacology of inhibitory neurotransmission better than ATP. Here ß-NAD and ATP were compared as possible inhibitory neurotransmitters in human and monkey colons. METHODS: A small-volume superfusion assay and high-pressure liquid chromatography with fluorescence detection were used to evaluate spontaneous and nerve-evoked overflow of ß-NAD, ATP, and metabolites. Postjunctional responses to nerve stimulation, ß-NAD and ATP were compared using intracellular membrane potential and force measurements. Effects of ß-NAD on smooth muscle cells (SMCs) were recorded by patch clamp. P2Y receptor transcripts were assayed by reverse transcription polymerase chain reaction. RESULTS: In contrast to ATP, overflow of ß-NAD evoked by electrical field stimulation correlated with stimulation frequency and was diminished by the neurotoxins, tetrodotoxin, and ω-conotoxin GVIA. Inhibitory junction potentials and responses to exogenous ß-NAD, but not ATP, were blocked by P2Y receptor antagonists suramin, pyridoxal-phosphate-6-azophenyl-2',4'-disulfonate (PPADS), 2'-deoxy-N6-methyladenosine 3',5'-bisphosphate (MRS 2179), and (1R,2S,4S,5S)-4-[2-Iodo-6-(methylamino)-9H-purin-9-yl]-2-(phosphonooxy)bicyclo[3.1.0]hexane-1-methanol dihydrogen phosphate ester tetraammonium salt (MRS 2500). ß-NAD activated nonselective cation currents in SMCs, but failed to activate outward currents. CONCLUSIONS: ß-NAD meets the criteria for a neurotransmitter better than ATP in human and monkey colons and therefore may contribute to neural regulation of colonic motility. SMCs are unlikely targets for inhibitory purine neurotransmitters because dominant responses of SMCs were activation of net inward, rather than outward, current.


Asunto(s)
Colon/inervación , Sistema Nervioso Entérico/fisiología , NAD/fisiología , Transmisión Sináptica/fisiología , Adenosina Trifosfato/análisis , Adenosina Trifosfato/antagonistas & inhibidores , Adenosina Trifosfato/farmacología , Adenosina Trifosfato/fisiología , Adulto , Anciano , Animales , Colon/efectos de los fármacos , Estimulación Eléctrica , Sistema Nervioso Entérico/efectos de los fármacos , Humanos , Canales Iónicos/efectos de los fármacos , Canales Iónicos/fisiología , Macaca , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Persona de Mediana Edad , Músculo Liso/efectos de los fármacos , Músculo Liso/inervación , Músculo Liso/fisiología , NAD/farmacología , Neurotoxinas/farmacología , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y/análisis , Transmisión Sináptica/efectos de los fármacos , Tetrodotoxina/farmacología , omega-Conotoxina GVIA/farmacología
18.
J Physiol ; 589(Pt 3): 697-710, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21173079

RESUMEN

Smooth muscles, as in the gastrointestinal tract, are composed of several types of cells. Gastrointestinal muscles contain smooth muscle cells, enteric neurons, glial cells, immune cells, and various classes of interstitial cells. One type of interstitial cell, referred to as 'fibroblast-like cells' by morphologists, are common, but their function is unknown. These cells are found near the terminals of enteric motor neurons, suggesting they could have a role in generating neural responses that help control gastrointestinal movements. We used a novel mouse with bright green fluorescent protein expressed specifically in the fibroblast-like cells to help us identify these cells in the mixture of cells obtained when whole muscles are dispersed with enzymes. We isolated these cells and found they respond to a major class of inhibitory neurotransmitters - purines. We characterized these responses, and our results provide a new hypothesis about the role of fibroblast-like cells in smooth muscle tissues.


Asunto(s)
Fibroblastos/citología , Fibroblastos/fisiología , Tracto Gastrointestinal/fisiología , Músculo Liso/fisiología , Transmisión Sináptica/fisiología , Adenosina Difosfato/farmacología , Adenosina Trifosfato/farmacología , Animales , Apamina/farmacología , Calcio/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/fisiología , Caribdotoxina/farmacología , Nucleótidos de Desoxiadenina/farmacología , Fenómenos Electrofisiológicos/efectos de los fármacos , Fenómenos Electrofisiológicos/fisiología , Fibroblastos/efectos de los fármacos , Tracto Gastrointestinal/citología , Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/fisiología , NAD/farmacología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Técnicas de Placa-Clamp , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores Purinérgicos P2Y1/genética , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Proteínas de Transporte Vesicular de Acetilcolina/metabolismo
19.
J Urol ; 183(2): 793-800, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20022044

RESUMEN

PURPOSE: Mouse models of partial bladder outlet obstruction cause bladder hypertrophy. Expression of a number of ion channels is altered in hypertrophic detrusor muscle, resulting in bladder dysfunction. We determined whether mechanosensitive TREK-1 channels are present in the murine bladder and whether their expression is altered in partial bladder outlet obstruction, resulting in abnormal filling responses. MATERIALS AND METHODS: Partial bladder outlet obstruction was surgically induced in CD-1 mice and the mice recovered for 14 days. Cystometry was done to evaluate bladder pressure responses during filling at 25 microl per minute in partial bladder outlet obstruction mice and sham operated controls. TREK-1 channel expression was determined at the mRNA and protein levels by quantitative reverse transcriptase-polymerase chain reaction and Western blotting, respectively, and localized in the bladder wall using immunohistochemistry. RESULTS: Obstructed bladders showed about a 2-fold increase in weight vs sham operated bladders. TREK-1 channel protein expression on Western blots from bladder smooth muscle strip homogenates was significantly decreased in obstructed mice. Immunohistochemistry revealed a significant decrease in TREK-1 channel immunoreactivity in detrusor smooth muscle in obstructed mice. On cystometry the TREK-1 channel blocker L-methioninol induced a significant increase in premature contractions during filling in sham operated mice. L-methioninol had no significant effect in obstructed mice, which showed an overactive detrusor phenotype. CONCLUSIONS: TREK-1 channel down-regulation in detrusor myocytes is associated with bladder overactivity in a murine model of partial bladder outlet obstruction.


Asunto(s)
Canales de Potasio de Dominio Poro en Tándem/fisiología , Obstrucción del Cuello de la Vejiga Urinaria/complicaciones , Vejiga Urinaria Hiperactiva/etiología , Animales , Femenino , Ratones , Vejiga Urinaria Hiperactiva/patología , Vejiga Urinaria Hiperactiva/fisiopatología
20.
J Physiol ; 587(Pt 20): 4905-18, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19703958

RESUMEN

Interstitial cells of Cajal (ICC) are unique cells that generate electrical pacemaker activity in gastrointestinal (GI) muscles. Many previous studies have attempted to characterize the conductances responsible for pacemaker current and slow waves in the GI tract, but the precise mechanism of electrical rhythmicity is still debated. We used a new transgenic mouse with a bright green fluorescent protein (copGFP) constitutively expressed in ICC to facilitate study of these cells in mixed cell dispersions. We found that ICC express a specialized 'slow wave' current. Reversal of tail current analysis showed this current was due to a Cl(-) selective conductance. ICC express ANO1, a Ca(2+)-activated Cl(-) channel. Slow wave currents are not voltage dependent, but a secondary voltage-dependent process underlies activation of these currents. Removal of extracellular Ca(2+), replacement of Ca(2+) with Ba(2+), or extracellular Ni(2+) (30 microm) blocked the slow wave current. Single Ca(2+)-activated Cl() channels with a unitary conductance of 7.8 pS were resolved in excised patches of ICC. These are similar in conductance to ANO1 channels (8 pS) expressed in HEK293 cells. Slow wave current was blocked in a concentration-dependent manner by niflumic acid (IC(50) = 4.8 microm). Slow wave currents are associated with transient depolarizations of ICC in current clamp, and these events were blocked by niflumic acid. These findings demonstrate a role for a Ca(2+)-activated Cl(-) conductance in slow wave current in ICC and are consistent with the idea that ANO1 participates in pacemaker activity.


Asunto(s)
Motilidad Gastrointestinal/fisiología , Células Intersticiales de Cajal/fisiología , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Animales , Anoctamina-1 , Calcio/metabolismo , Canales de Cloruro , Inhibidores de la Ciclooxigenasa/farmacología , Femenino , Motilidad Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/fisiología , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes , Células Intersticiales de Cajal/citología , Células Intersticiales de Cajal/efectos de los fármacos , Ratones , Músculo Liso/citología , Músculo Liso/fisiología , Ácido Niflúmico/farmacología , Técnicas de Placa-Clamp , Periodicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA