Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(16): 3386-3399.e15, 2023 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-37541196

RESUMEN

The gastrointestinal tract is in a state of constant motion. These movements are tightly regulated by the presence of food and help digestion by mechanically breaking down and propelling gut content. Mechanical sensing in the gut is thought to be essential for regulating motility; however, the identity of the neuronal populations, the molecules involved, and the functional consequences of this sensation are unknown. Here, we show that humans lacking PIEZO2 exhibit impaired bowel sensation and motility. Piezo2 in mouse dorsal root, but not nodose ganglia is required to sense gut content, and this activity slows down food transit rates in the stomach, small intestine, and colon. Indeed, Piezo2 is directly required to detect colon distension in vivo. Our study unveils the mechanosensory mechanisms that regulate the transit of luminal contents throughout the gut, which is a critical process to ensure proper digestion, nutrient absorption, and waste removal.


Asunto(s)
Tránsito Gastrointestinal , Canales Iónicos , Mecanotransducción Celular , Animales , Humanos , Ratones , Digestión , Canales Iónicos/metabolismo , Neuronas/metabolismo
2.
Cell ; 184(4): 969-982.e13, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33571427

RESUMEN

Iron overload causes progressive organ damage and is associated with arthritis, liver damage, and heart failure. Elevated iron levels are present in 1%-5% of individuals; however, iron overload is undermonitored and underdiagnosed. Genetic factors affecting iron homeostasis are emerging. Individuals with hereditary xerocytosis, a rare disorder with gain-of-function (GOF) mutations in mechanosensitive PIEZO1 ion channel, develop age-onset iron overload. We show that constitutive or macrophage expression of a GOF Piezo1 allele in mice disrupts levels of the iron regulator hepcidin and causes iron overload. We further show that PIEZO1 is a key regulator of macrophage phagocytic activity and subsequent erythrocyte turnover. Strikingly, we find that E756del, a mild GOF PIEZO1 allele present in one-third of individuals of African descent, is strongly associated with increased plasma iron. Our study links macrophage mechanotransduction to iron metabolism and identifies a genetic risk factor for increased iron levels in African Americans.


Asunto(s)
Canales Iónicos/metabolismo , Hierro/metabolismo , Negro o Afroamericano , Envejecimiento/metabolismo , Alelos , Animales , Estudios de Cohortes , Recuento de Eritrocitos , Eritropoyesis , Mutación con Ganancia de Función/genética , Hepatocitos/metabolismo , Hepcidinas/sangre , Hepcidinas/metabolismo , Humanos , Hierro/sangre , Sobrecarga de Hierro/metabolismo , Macrófagos/metabolismo , Mecanotransducción Celular , Ratones Endogámicos C57BL , Fagocitosis , Fenotipo , Estrés Fisiológico
3.
Cell ; 173(2): 443-455.e12, 2018 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-29576450

RESUMEN

Hereditary xerocytosis is thought to be a rare genetic condition characterized by red blood cell (RBC) dehydration with mild hemolysis. RBC dehydration is linked to reduced Plasmodium infection in vitro; however, the role of RBC dehydration in protection against malaria in vivo is unknown. Most cases of hereditary xerocytosis are associated with gain-of-function mutations in PIEZO1, a mechanically activated ion channel. We engineered a mouse model of hereditary xerocytosis and show that Plasmodium infection fails to cause experimental cerebral malaria in these mice due to the action of Piezo1 in RBCs and in T cells. Remarkably, we identified a novel human gain-of-function PIEZO1 allele, E756del, present in a third of the African population. RBCs from individuals carrying this allele are dehydrated and display reduced Plasmodium infection in vitro. The existence of a gain-of-function PIEZO1 at such high frequencies is surprising and suggests an association with malaria resistance.


Asunto(s)
Anemia Hemolítica Congénita/patología , Población Negra/genética , Hidropesía Fetal/patología , Canales Iónicos/genética , Malaria/patología , Alelos , Anemia Hemolítica Congénita/genética , Animales , Deshidratación , Modelos Animales de Enfermedad , Eritrocitos/citología , Eritrocitos/metabolismo , Eliminación de Gen , Genotipo , Humanos , Hidropesía Fetal/genética , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/deficiencia , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Canales Iónicos/química , Malaria/genética , Malaria/parasitología , Malaria/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Plasmodium berghei/crecimiento & desarrollo , Plasmodium berghei/patogenicidad , Linfocitos T/citología , Linfocitos T/metabolismo
4.
Cell ; 173(3): 762-775.e16, 2018 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-29677517

RESUMEN

Mechanotransduction plays a crucial role in vascular biology. One example of this is the local regulation of vascular resistance via flow-mediated dilation (FMD). Impairment of this process is a hallmark of endothelial dysfunction and a precursor to a wide array of vascular diseases, such as hypertension and atherosclerosis. Yet the molecules responsible for sensing flow (shear stress) within endothelial cells remain largely unknown. We designed a 384-well screening system that applies shear stress on cultured cells. We identified a mechanosensitive cell line that exhibits shear stress-activated calcium transients, screened a focused RNAi library, and identified GPR68 as necessary and sufficient for shear stress responses. GPR68 is expressed in endothelial cells of small-diameter (resistance) arteries. Importantly, Gpr68-deficient mice display markedly impaired acute FMD and chronic flow-mediated outward remodeling in mesenteric arterioles. Therefore, GPR68 is an essential flow sensor in arteriolar endothelium and is a critical signaling component in cardiovascular pathophysiology.


Asunto(s)
Mecanotransducción Celular , Interferencia de ARN , Receptores Acoplados a Proteínas G/fisiología , Animales , Materiales Biocompatibles , Calcio/metabolismo , Línea Celular Tumoral , Células Endoteliales/fisiología , Endotelio Vascular/citología , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Concentración de Iones de Hidrógeno , Arterias Mesentéricas/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo , ARN Interferente Pequeño/metabolismo , Receptores Acoplados a Proteínas G/genética , Resistencia al Corte , Estrés Mecánico , Resistencia Vascular
5.
Cell ; 164(3): 499-511, 2016 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-26824658

RESUMEN

The volume-regulated anion channel (VRAC) is activated when a cell swells, and it plays a central role in maintaining cell volume in response to osmotic challenges. SWELL1 (LRRC8A) was recently identified as an essential component of VRAC. However, the identity of the pore-forming subunits of VRAC and how the channel is gated by cell swelling are unknown. Here, we show that SWELL1 and up to four other LRRC8 subunits assemble into heterogeneous complexes of ∼800 kDa. When reconstituted into bilayers, LRRC8 complexes are sufficient to form anion channels activated by osmolality gradients. In bilayers, as well as in cells, the single-channel conductance of the complexes depends on the LRRC8 composition. Finally, low ionic strength (Γ) in the absence of an osmotic gradient activates the complexes in bilayers. These data demonstrate that LRRC8 proteins together constitute the VRAC pore and that hypotonic stress can activate VRAC through a decrease in cytoplasmic Γ.


Asunto(s)
Canales Iónicos/metabolismo , Proteínas de la Membrana/metabolismo , Células HeLa , Humanos , Canales Iónicos/química , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Ósmosis
6.
Nat Rev Mol Cell Biol ; 18(12): 771-783, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28974772

RESUMEN

Cellular mechanotransduction, the process of translating mechanical forces into biological signals, is crucial for a wide range of physiological processes. A role for ion channels in sensing mechanical forces has been proposed for decades, but their identity in mammals remained largely elusive until the discovery of Piezos. Recent research on Piezos has underscored their importance in somatosensation (touch perception, proprioception and pulmonary respiration), red blood cell volume regulation, vascular physiology and various human genetic disorders.


Asunto(s)
Enfermedades Genéticas Congénitas/metabolismo , Activación del Canal Iónico , Canales Iónicos/metabolismo , Propiocepción , Mecánica Respiratoria , Percepción del Tacto , Animales , Enfermedades Genéticas Congénitas/genética , Humanos , Canales Iónicos/genética
7.
Cell ; 157(2): 447-458, 2014 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-24725410

RESUMEN

Maintenance of a constant cell volume in response to extracellular or intracellular osmotic changes is critical for cellular homeostasis. Activation of a ubiquitous volume-regulated anion channel (VRAC) plays a key role in this process; however, its molecular identity in vertebrates remains unknown. Here, we used a cell-based fluorescence assay and performed a genome-wide RNAi screen to find components of VRAC. We identified SWELL1 (LRRC8A), a member of a four-transmembrane protein family with unknown function, as essential for hypotonicity-induced iodide influx. SWELL1 is localized to the plasma membrane, and its knockdown dramatically reduces endogenous VRAC currents and regulatory cell volume decrease in various cell types. Furthermore, point mutations in SWELL1 cause a significant change in VRAC anion selectivity, demonstrating that SWELL1 is an essential VRAC component. These findings enable further molecular characterization of the VRAC channel complex and genetic studies for understanding the function of VRAC in normal physiology and disease.


Asunto(s)
Tamaño de la Célula , Proteínas de la Membrana/metabolismo , Animales , Membrana Celular/química , Membrana Celular/metabolismo , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Estudio de Asociación del Genoma Completo , Células HEK293 , Células HeLa , Humanos , Yoduros/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Interferencia de ARN
8.
Nature ; 620(7976): 1117-1125, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37587339

RESUMEN

PIEZOs are mechanosensitive ion channels that convert force into chemoelectric signals1,2 and have essential roles in diverse physiological settings3. In vitro studies have proposed that PIEZO channels transduce mechanical force through the deformation of extensive blades of transmembrane domains emanating from a central ion-conducting pore4-8. However, little is known about how these channels interact with their native environment and which molecular movements underlie activation. Here we directly observe the conformational dynamics of the blades of individual PIEZO1 molecules in a cell using nanoscopic fluorescence imaging. Compared with previous structural models of PIEZO1, we show that the blades are significantly expanded at rest by the bending stress exerted by the plasma membrane. The degree of expansion varies dramatically along the length of the blade, where decreased binding strength between subdomains can explain increased flexibility of the distal blade. Using chemical and mechanical modulators of PIEZO1, we show that blade expansion and channel activation are correlated. Our findings begin to uncover how PIEZO1 is activated in a native environment. More generally, as we reliably detect conformational shifts of single nanometres from populations of channels, we expect that this approach will serve as a framework for the structural analysis of membrane proteins through nanoscopic imaging.


Asunto(s)
Canales Iónicos , Membrana Celular/metabolismo , Fluorescencia , Canales Iónicos/química , Canales Iónicos/metabolismo , Modelos Moleculares , Movimiento , Conformación Proteica , Análisis de la Célula Individual
9.
Nature ; 607(7917): 104-110, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35732741

RESUMEN

Itch triggers scratching, a behavioural defence mechanism that aids in the removal of harmful irritants and parasites1. Chemical itch is triggered by many endogenous and exogenous cues, such as pro-inflammatory histamine, which is released during an allergic reaction1. Mechanical itch can be triggered by light sensations such as wool fibres or a crawling insect2. In contrast to chemical itch pathways, which have been extensively studied, the mechanisms that underlie the transduction of mechanical itch are largely unknown. Here we show that the mechanically activated ion channel PIEZO1 (ref. 3) is selectively expressed by itch-specific sensory neurons and is required for their mechanically activated currents. Loss of PIEZO1 function in peripheral neurons greatly reduces mechanically evoked scratching behaviours and both acute and chronic itch-evoked sensitization. Finally, mice expressing a gain-of-function Piezo1 allele4 exhibit enhanced mechanical itch behaviours. Our studies reveal the polymodal nature of itch sensory neurons and identify a role for PIEZO1 in the sensation of itch.


Asunto(s)
Canales Iónicos , Prurito , Alelos , Animales , Canales Iónicos/deficiencia , Canales Iónicos/genética , Canales Iónicos/metabolismo , Ratones , Prurito/genética , Prurito/fisiopatología , Sensación , Células Receptoras Sensoriales/metabolismo
10.
Nature ; 609(7927): 569-574, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36045288

RESUMEN

Adipose tissues communicate with the central nervous system to maintain whole-body energy homeostasis. The mainstream view is that circulating hormones secreted by the fat convey the metabolic state to the brain, which integrates peripheral information and regulates adipocyte function through noradrenergic sympathetic output1. Moreover, somatosensory neurons of the dorsal root ganglia innervate adipose tissue2. However, the lack of genetic tools to selectively target these neurons has limited understanding of their physiological importance. Here we developed viral, genetic and imaging strategies to manipulate sensory nerves in an organ-specific manner in mice. This enabled us to visualize the entire axonal projection of dorsal root ganglia from the soma to subcutaneous adipocytes, establishing the anatomical underpinnings of adipose sensory innervation. Functionally, selective sensory ablation in adipose tissue enhanced the lipogenic and thermogenetic transcriptional programs, resulting in an enlarged fat pad, enrichment of beige adipocytes and elevated body temperature under thermoneutral conditions. The sensory-ablation-induced phenotypes required intact sympathetic function. We postulate that beige-fat-innervating sensory neurons modulate adipocyte function by acting as a brake on the sympathetic system. These results reveal an important role of the innervation by dorsal root ganglia of adipose tissues, and could enable future studies to examine the role of sensory innervation of disparate interoceptive systems.


Asunto(s)
Tejido Adiposo , Células Receptoras Sensoriales , Tejido Adiposo/inervación , Tejido Adiposo/metabolismo , Tejido Adiposo Beige/inervación , Tejido Adiposo Beige/metabolismo , Animales , Axones , Metabolismo Energético , Ganglios Espinales/fisiología , Homeostasis , Hormonas/metabolismo , Ratones , Especificidad de Órganos , Células Receptoras Sensoriales/fisiología , Grasa Subcutánea/inervación , Grasa Subcutánea/metabolismo , Sistema Nervioso Simpático/citología , Sistema Nervioso Simpático/fisiología , Termogénesis/genética
11.
Cell ; 147(2): 261-2, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-22000005

RESUMEN

Opioids such as morphine numb pain but often concomitantly induce itch. Liu et al. (2011) now separate the sensation of itch from opioid-induced analgesia, showing that in a subset of spinal neurons, morphine directly induces itch by signaling through a heteromer of opioid- and itch-mediating G protein-coupled receptors.

12.
Nature ; 588(7837): 290-295, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33057202

RESUMEN

Henry Miller stated that "to relieve a full bladder is one of the great human joys". Urination is critically important in health and ailments of the lower urinary tract cause high pathological burden. Although there have been advances in understanding the central circuitry in the brain that facilitates urination1-3, there is a lack of in-depth mechanistic insight into the process. In addition to central control, micturition reflexes that govern urination are all initiated by peripheral mechanical stimuli such as bladder stretch and urethral flow4. The mechanotransduction molecules and cell types that function as the primary stretch and pressure detectors in the urinary tract mostly remain unknown. Here we identify expression of the mechanosensitive ion channel PIEZO2 in lower urinary tract tissues, where it is required for low-threshold bladder-stretch sensing and urethral micturition reflexes. We show that PIEZO2 acts as a sensor in both the bladder urothelium and innervating sensory neurons. Humans and mice lacking functional PIEZO2 have impaired bladder control, and humans lacking functional PIEZO2 report deficient bladder-filling sensation. This study identifies PIEZO2 as a key mechanosensor in urinary function. These findings set the foundation for future work to identify the interactions between urothelial cells and sensory neurons that control urination.


Asunto(s)
Canales Iónicos/metabolismo , Mecanotransducción Celular/fisiología , Células Receptoras Sensoriales/metabolismo , Vejiga Urinaria/inervación , Vejiga Urinaria/fisiología , Micción/fisiología , Urotelio/citología , Animales , Femenino , Humanos , Canales Iónicos/deficiencia , Ratones , Presión , Reflejo/fisiología , Vejiga Urinaria/citología , Vejiga Urinaria/fisiopatología , Sistema Urinario/inervación , Sistema Urinario/metabolismo , Urotelio/metabolismo
13.
Cell ; 139(7): 1224-6, 2009 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-20064368

RESUMEN

The itch sensation results from the excitation of primary sensory nerve endings in the skin, but the underlying molecular mechanisms are not completely understood. Liu et al. (2009) now report that some members of the Mrgpr class of G protein-coupled receptors mediate the itch caused by the antimalarial drug chloroquine.


Asunto(s)
Prurito/fisiopatología , Animales , Antimaláricos/efectos adversos , Cloroquina/efectos adversos , Humanos , Dolor/fisiopatología , Células Receptoras Sensoriales/fisiología , Piel/inervación
14.
Nature ; 554(7693): 481-486, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29261642

RESUMEN

Piezo1 and Piezo2 are mechanically activated ion channels that mediate touch perception, proprioception and vascular development. Piezo proteins are distinct from other ion channels and their structure remains poorly defined, which impedes detailed study of their gating and ion permeation properties. Here we report a high-resolution cryo-electron microscopy structure of the mouse Piezo1 trimer. The detergent-solubilized complex adopts a three-bladed propeller shape with a curved transmembrane region containing at least 26 transmembrane helices per protomer. The flexible propeller blades can adopt distinct conformations, and consist of a series of four-transmembrane helical bundles that we term Piezo repeats. Carboxy-terminal domains line the central ion pore, and the channel is closed by constrictions in the cytosol. A kinked helical beam and anchor domain link the Piezo repeats to the pore, and are poised to control gating allosterically. The structure provides a foundation to dissect further how Piezo channels are regulated by mechanical force.


Asunto(s)
Microscopía por Crioelectrón , Canales Iónicos/química , Canales Iónicos/ultraestructura , Animales , Sitios de Unión , Activación del Canal Iónico , Canales Iónicos/genética , Canales Iónicos/metabolismo , Lípidos , Ratones , Modelos Moleculares , Mutación , Docilidad , Dominios Proteicos , Solubilidad
15.
Proc Natl Acad Sci U S A ; 118(20)2021 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-33975957

RESUMEN

Plant roots adapt to the mechanical constraints of the soil to grow and absorb water and nutrients. As in animal species, mechanosensitive ion channels in plants are proposed to transduce external mechanical forces into biological signals. However, the identity of these plant root ion channels remains unknown. Here, we show that Arabidopsis thaliana PIEZO1 (PZO1) has preserved the function of its animal relatives and acts as an ion channel. We present evidence that plant PIEZO1 is expressed in the columella and lateral root cap cells of the root tip, which are known to experience robust mechanical strain during root growth. Deleting PZO1 from the whole plant significantly reduced the ability of its roots to penetrate denser barriers compared to wild-type plants. pzo1 mutant root tips exhibited diminished calcium transients in response to mechanical stimulation, supporting a role of PZO1 in root mechanotransduction. Finally, a chimeric PZO1 channel that includes the C-terminal half of PZO1 containing the putative pore region was functional and mechanosensitive when expressed in naive mammalian cells. Collectively, our data suggest that Arabidopsis PIEZO1 plays an important role in root mechanotransduction and establish PIEZOs as physiologically relevant mechanosensitive ion channels across animal and plant kingdoms.


Asunto(s)
Proteínas de Arabidopsis/fisiología , Arabidopsis/fisiología , Mecanotransducción Celular/fisiología , Proteínas de Transporte de Membrana/fisiología , Raíces de Plantas/fisiología
17.
Nature ; 541(7636): 176-181, 2017 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-28002412

RESUMEN

Respiratory dysfunction is a notorious cause of perinatal mortality in infants and sleep apnoea in adults, but the mechanisms of respiratory control are not clearly understood. Mechanical signals transduced by airway-innervating sensory neurons control respiration; however, the physiological significance and molecular mechanisms of these signals remain obscured. Here we show that global and sensory neuron-specific ablation of the mechanically activated ion channel Piezo2 causes respiratory distress and death in newborn mice. Optogenetic activation of Piezo2+ vagal sensory neurons causes apnoea in adult mice. Moreover, induced ablation of Piezo2 in sensory neurons of adult mice causes decreased neuronal responses to lung inflation, an impaired Hering-Breuer mechanoreflex, and increased tidal volume under normal conditions. These phenotypes are reproduced in mice lacking Piezo2 in the nodose ganglion. Our data suggest that Piezo2 is an airway stretch sensor and that Piezo2-mediated mechanotransduction within various airway-innervating sensory neurons is critical for establishing efficient respiration at birth and maintaining normal breathing in adults.


Asunto(s)
Apnea/fisiopatología , Canales Iónicos/metabolismo , Pulmón/fisiología , Pulmón/fisiopatología , Mecanotransducción Celular/fisiología , Reflejo/fisiología , Animales , Animales Recién Nacidos , Apnea/genética , Muerte , Femenino , Canales Iónicos/deficiencia , Canales Iónicos/genética , Masculino , Mecanotransducción Celular/genética , Ratones , Ganglio Nudoso/metabolismo , Reflejo/genética , Respiración , Células Receptoras Sensoriales/metabolismo , Volumen de Ventilación Pulmonar
18.
Am J Hum Genet ; 105(5): 996-1004, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31587869

RESUMEN

Mechanically activated (MA) ion channels convert physical forces into electrical signals. Despite the importance of this function, the involvement of mechanosensitive ion channels in human disease is poorly understood. Here we report heterozygous missense mutations in the gene encoding the MA ion channel TMEM63A that result in an infantile disorder resembling a hypomyelinating leukodystrophy. Four unrelated individuals presented with congenital nystagmus, motor delay, and deficient myelination on serial scans in infancy, prompting the diagnosis of Pelizaeus-Merzbacher (like) disease. Genomic sequencing revealed that all four individuals carry heterozygous missense variants in the pore-forming domain of TMEM63A. These variants were confirmed to have arisen de novo in three of the four individuals. While the physiological role of TMEM63A is incompletely understood, it is highly expressed in oligodendrocytes and it has recently been shown to be a MA ion channel. Using patch clamp electrophysiology, we demonstrated that each of the modeled variants result in strongly attenuated stretch-activated currents when expressed in naive cells. Unexpectedly, the clinical evolution of all four individuals has been surprisingly favorable, with substantial improvements in neurological signs and developmental progression. In the three individuals with follow-up scans after 4 years of age, the myelin deficit had almost completely resolved. Our results suggest a previously unappreciated role for mechanosensitive ion channels in myelin development.


Asunto(s)
Canales Iónicos/genética , Proteínas de la Membrana/genética , Vaina de Mielina/genética , Enfermedad de Pelizaeus-Merzbacher/genética , Adolescente , Adulto , Preescolar , Femenino , Heterocigoto , Humanos , Masculino , Mutación Missense/genética , Oligodendroglía/metabolismo , Adulto Joven
19.
Proc Natl Acad Sci U S A ; 115(50): 12817-12822, 2018 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-30482854

RESUMEN

PIEZO1 is a cation channel that is activated by mechanical forces such as fluid shear stress or membrane stretch. PIEZO1 loss-of-function mutations in patients are associated with congenital lymphedema with pleural effusion. However, the mechanistic link between PIEZO1 function and the development or function of the lymphatic system is currently unknown. Here, we analyzed two mouse lines lacking PIEZO1 in endothelial cells (via Tie2Cre or Lyve1Cre) and found that they exhibited pleural effusion and died postnatally. Strikingly, the number of lymphatic valves was dramatically reduced in these mice. Lymphatic valves are essential for ensuring proper circulation of lymph. Mechanical forces have been implicated in the development of lymphatic vasculature and valve formation, but the identity of mechanosensors involved is unknown. Expression of FOXC2 and NFATc1, transcription factors known to be required for lymphatic valve development, appeared normal in Tie2Cre;Piezo1cKO mice. However, the process of protrusion in the valve leaflets, which is associated with collective cell migration, actin polymerization, and remodeling of cell-cell junctions, was impaired in Tie2Cre;Piezo1cKO mice. Consistent with these genetic findings, activation of PIEZO1 by Yoda1 in cultured lymphatic endothelial cells induced active remodeling of actomyosin and VE-cadherin+ cell-cell adhesion sites. Our analysis provides evidence that mechanically activated ion channel PIEZO1 is a key regulator of lymphatic valve formation.


Asunto(s)
Canales Iónicos/metabolismo , Linfangiogénesis/fisiología , Sistema Linfático/metabolismo , Sistema Linfático/fisiología , Vasos Linfáticos/metabolismo , Vasos Linfáticos/fisiología , Actomiosina/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Células Endoteliales/metabolismo , Células Endoteliales/fisiología , Factores de Transcripción Forkhead/metabolismo , Uniones Intercelulares/metabolismo , Uniones Intercelulares/fisiología , Transporte Iónico/fisiología , Ratones , Factores de Transcripción NFATC/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo
20.
Nature ; 509(7502): 622-6, 2014 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-24717433

RESUMEN

How we sense touch remains fundamentally unknown. The Merkel cell-neurite complex is a gentle touch receptor in the skin that mediates slowly adapting responses of Aß sensory fibres to encode fine details of objects. This mechanoreceptor complex was recognized to have an essential role in sensing gentle touch nearly 50 years ago. However, whether Merkel cells or afferent fibres themselves sense mechanical force is still debated, and the molecular mechanism of mechanotransduction is unknown. Synapse-like junctions are observed between Merkel cells and associated afferents, and yet it is unclear whether Merkel cells are inherently mechanosensitive or whether they can rapidly transmit such information to the neighbouring nerve. Here we show that Merkel cells produce touch-sensitive currents in vitro. Piezo2, a mechanically activated cation channel, is expressed in Merkel cells. We engineered mice deficient in Piezo2 in the skin, but not in sensory neurons, and show that Merkel-cell mechanosensitivity completely depends on Piezo2. In these mice, slowly adapting responses in vivo mediated by the Merkel cell-neurite complex show reduced static firing rates, and moreover, the mice display moderately decreased behavioural responses to gentle touch. Our results indicate that Piezo2 is the Merkel-cell mechanotransduction channel and provide the first line of evidence that Piezo channels have a physiological role in mechanosensation in mammals. Furthermore, our data present evidence for a two-receptor-site model, in which both Merkel cells and innervating afferents act together as mechanosensors. The two-receptor system could provide this mechanoreceptor complex with a tuning mechanism to achieve highly sophisticated responses to a given mechanical stimulus.


Asunto(s)
Canales Iónicos/metabolismo , Mecanotransducción Celular , Células de Merkel/metabolismo , Tacto/fisiología , Potenciales de Acción , Animales , Conductividad Eléctrica , Femenino , Técnicas In Vitro , Canales Iónicos/deficiencia , Canales Iónicos/genética , Masculino , Mecanotransducción Celular/genética , Ratones , Ratones Noqueados , Neuritas/metabolismo , Neuronas Aferentes/metabolismo , Piel/citología , Piel/inervación , Tacto/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA