Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
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
2.
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
3.
Proc Natl Acad Sci U S A ; 111(28): 10347-52, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24958852

RESUMEN

Mechanosensation is perhaps the last sensory modality not understood at the molecular level. Ion channels that sense mechanical force are postulated to play critical roles in a variety of biological processes including sensing touch/pain (somatosensation), sound (hearing), and shear stress (cardiovascular physiology); however, the identity of these ion channels has remained elusive. We previously identified Piezo1 and Piezo2 as mechanically activated cation channels that are expressed in many mechanosensitive cell types. Here, we show that Piezo1 is expressed in endothelial cells of developing blood vessels in mice. Piezo1-deficient embryos die at midgestation with defects in vascular remodeling, a process critically influenced by blood flow. We demonstrate that Piezo1 is activated by shear stress, the major type of mechanical force experienced by endothelial cells in response to blood flow. Furthermore, loss of Piezo1 in endothelial cells leads to deficits in stress fiber and cellular orientation in response to shear stress, linking Piezo1 mechanotransduction to regulation of cell morphology. These findings highlight an essential role of mammalian Piezo1 in vascular development during embryonic development.


Asunto(s)
Sistema Cardiovascular/embriología , Desarrollo Embrionario/fisiología , Células Endoteliales/metabolismo , Canales Iónicos/metabolismo , Mecanotransducción Celular/fisiología , Animales , Sistema Cardiovascular/citología , Células Endoteliales/citología , Canales Iónicos/genética , Ratones , Ratones Transgénicos
4.
Mol Pharmacol ; 89(1): 176-86, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26494861

RESUMEN

The molecular pharmacology of the G protein-coupled receptors for sphingosine 1-phosphate (S1P) provides important insight into established and new therapeutic targets. A new, potent bitopic S1P3 antagonist, SPM-354, with in vivo activity, has been used, together with S1P3-knockin and S1P3-knockout mice to define the spatial and functional properties of S1P3 in regulating cardiac conduction. We show that S1P3 is a key direct regulator of cardiac rhythm both in vivo and in isolated perfused hearts. 2-Amino-2-[2-(4-octylphenyl)ethyl]propane-1,3-diol in vivo and S1P in isolated hearts induced a spectrum of cardiac effects, ranging from sinus bradycardia to complete heart block, as measured by a surface electrocardiogram in anesthetized mice and in volume-conducted Langendorff preparations. The agonist effects on complete heart block are absent in S1P3-knockout mice and are reversed in wild-type mice with SPM-354, as characterized and described here. Homologous knockin of S1P3-mCherry is fully functional pharmacologically and is strongly expressed by immunohistochemistry confocal microscopy in Hyperpolarization Activated Cyclic Nucleotide Gated Potassium Channel 4 (HCN4)-positive atrioventricular node and His-Purkinje fibers, with relative less expression in the HCN4-positive sinoatrial node. In Langendorff studies, at constant pressure, SPM-354 restored sinus rhythm in S1P-induced complete heart block and fully reversed S1P-mediated bradycardia. S1P3 distribution and function in the mouse ventricular cardiac conduction system suggest a direct mechanism for heart block risk that should be further studied in humans. A richer understanding of receptor and ligand usage in the pacemaker cells of the cardiac system is likely to be useful in understanding ventricular conduction in health, disease, and pharmacology.


Asunto(s)
Bloqueo Cardíaco/tratamiento farmacológico , Bloqueo Cardíaco/genética , Frecuencia Cardíaca/efectos de los fármacos , Frecuencia Cardíaca/fisiología , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/genética , Animales , Cardiotónicos/farmacología , Cardiotónicos/uso terapéutico , Bloqueo Cardíaco/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Esfingosina-1-Fosfato
5.
Curr Top Microbiol Immunol ; 378: 55-83, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24728593

RESUMEN

The zwitterionic lysophospholipid Sphingosine 1-Phosphate (S1P) is a pleiotropic mediator of physiology and pathology. The synthesis, transport, and degradation of S1P are tightly regulated to ensure that S1P is present in the proper concentrations in the proper location. The binding of S1P to five G protein-coupled S1P receptors regulates many physiological systems, particularly the immune and vascular systems. Our understanding of the functions of S1P has been aided by the tractability of the system to both chemical and genetic manipulation. Chemical modulators have been generated to affect most of the known components of S1P biology, including agonists of S1P receptors and inhibitors of enzymes regulating S1P production and degradation. Genetic knockouts and manipulations have been similarly engineered to disrupt the functions of individual S1P receptors or enzymes involved in S1P metabolism. This chapter will focus on the development and utilization of these chemical and genetic tools to explore the complex biology surrounding S1P and its receptors, with particular attention paid to the in vivo findings that these tools have allowed for.


Asunto(s)
Receptores de Lisoesfingolípidos/química , Receptores de Lisoesfingolípidos/genética , Animales , Técnicas Genéticas , Humanos , Lisofosfolípidos/química , Lisofosfolípidos/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/química , Esfingosina/metabolismo
6.
Mol Pharmacol ; 83(2): 316-21, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23204443

RESUMEN

Sphingosine 1-phosphate receptor 1 (S1P(1)) is a G protein-coupled receptor that is critical for proper lymphocyte development and recirculation. Agonists to S1P(1) are currently in use clinically for the treatment of multiple sclerosis, and these drugs may act on both S1P(1) expressed on lymphocytes and S1P(1) expressed within the central nervous system. Agonists to S1P(1) and deficiency in S1P(1) both cause lymphocyte sequestration in the lymph nodes. In the present study, we show that S1P(1) antagonism induces lymphocyte sequestration in the lymph nodes similar to that observed with S1P(1) agonists while upregulating S1P(1) on lymphocytes and endothelial cells. Additionally, we show that S1P(1) antagonism reverses experimental autoimmune encephalomyelitis in mice without acting on S1P(1) expressed within the central nervous system, demonstrating that lymphocyte sequestration via S1P(1) antagonism is sufficient to alleviate autoimmune pathology.


Asunto(s)
Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/genética , Inmunosupresores/farmacología , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/genética , Animales , Células CHO , Línea Celular , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/metabolismo , Cricetinae , Encefalomielitis Autoinmune Experimental/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Células HEK293 , Humanos , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Lisoesfingolípidos/metabolismo , Regulación hacia Arriba/efectos de los fármacos
7.
Nat Chem Biol ; 7(5): 254-6, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21445057

RESUMEN

Sphingosine 1-phosphate receptor 1 (S1P(1)) is critical for lymphocyte recirculation and is a clinical target for treatment of multiple sclerosis. By generating a short-duration S1P(1) agonist and mice in which fluorescently tagged S1P(1) replaces wild-type receptor, we elucidate physiological and agonist-perturbed changes in expression of S1P(1) at a subcellular level in vivo. We demonstrate differential downregulation of S1P(1) on lymphocytes and endothelia after agonist treatment.


Asunto(s)
Técnicas de Sustitución del Gen , Proteínas Fluorescentes Verdes/química , Esclerosis Múltiple/tratamiento farmacológico , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/uso terapéutico , Animales , Regulación hacia Abajo/efectos de los fármacos , Endotelio/efectos de los fármacos , Endotelio/metabolismo , Citometría de Flujo , Colorantes Fluorescentes/química , Colorantes Fluorescentes/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Ratones , Esclerosis Múltiple/metabolismo , Esclerosis Múltiple/patología , Receptores de Lisoesfingolípidos/metabolismo , Factores de Tiempo
8.
Am J Physiol Cell Physiol ; 302(10): C1460-8, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22357735

RESUMEN

Lymph nodes are highly organized structures specialized for efficient regulation of adaptive immunity. The blood and lymphatic systems within a lymph node play essential roles by providing functionally distinct environments for lymphocyte entry and egress, respectively. Direct imaging and measurement of vascular microenvironments by intravital multiphoton microscopy provide anatomical and mechanistic insights into the essential events of lymphocyte trafficking. Lymphocytes, blood endothelial cells, and lymphatic endothelial cells express sphingosine 1-phosphate receptor 1, a key G protein-coupled receptor regulating cellular egress and a modulator of endothelial permeability. Here we report the development of a differential vascular labeling (DVL) technique in which a single intravenous injection of a fluorescent dextran, in combination with fluorescent semiconductor quantum dot particles, differentially labels multiple blood and lymphatic compartments in a manner dependent on the size of the fluorescent particle used. Thus DVL allows measurement of endothelial integrity in multiple vascular compartments and the affects or pharmacological manipulation in vascular integrity. In addition, this technique allows for real-time observation of lymphocyte trafficking across physiological barriers differentiated by DVL. Last, single-field fluid movement dynamics can be derived, allowing for the simultaneous determination of fluid flow rates in diverse blood and lymphatic compartments.


Asunto(s)
Sistemas de Computación , Células Endoteliales/fisiología , Líquido Extracelular/química , Vasos Linfáticos/química , Flujo Sanguíneo Regional , Coloración y Etiquetado/métodos , Animales , Células Endoteliales/química , Líquido Extracelular/fisiología , Ganglios Linfáticos/irrigación sanguínea , Ganglios Linfáticos/química , Ganglios Linfáticos/fisiología , Vasos Linfáticos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Lisoesfingolípidos/biosíntesis , Receptores de Lisoesfingolípidos/sangre , Flujo Sanguíneo Regional/fisiología
9.
Mol Pharmacol ; 81(2): 166-74, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22031473

RESUMEN

Multiple sclerosis (MS) therapies modulate T-cell autoimmunity in the central nervous system (CNS) but may exacerbate latent infections. Fingolimod, a nonselective sphingosine-1-phosphate (S1P) receptor agonist that induces sustained lymphopenia and accumulates in the CNS, represents a new treatment modality for MS. We hypothesized that sustained lymphopenia would not be required for efficacy and that a selective, CNS-penetrant, peripherally short-acting, S1P(1) agonist would show full efficacy in a mouse MS model. Using daily treatment with 10 mg/kg 2-(4-(5-(3,4-diethoxyphenyl)-1,2,4-oxadiazol-3-yl)-2,3-dihydro-1H-inden-1-yl amino)ethanol (CYM-5442) at the onset of clinical signs in myelin oligodendrocyte glycoprotein MOG(35-55)- induced experimental allergic encephalomyelitis (EAE), we assessed clinical scores, CNS cellular infiltration, demyelination, and gliosis for 12 days with CYM-5442, vehicle, or fingolimod. CYM-5442 levels in CNS and plasma were determined at experiment termination, and blood lymphopenia was measured 3 and 24 h after the last injection. Plasma levels of cytokines were assayed at the end of the protocol. Changes in S1P(1)-enhanced green fluorescent protein expression on neurons and astrocytes during active EAE and upon CYM-5442 treatment were quantified with flow cytometry and Western blotting by using native-locus enhanced green fluorescent protein-tagged S1P(1) mice. S1P(1) agonism alone reduced pathological features as did fingolimod (maximally lymphopenic throughout), despite full reversal of lymphopenia within each dosing interval. CYM-5442 levels in CNS but not in plasma were sustained. Neuronal and astrocytic S1P(1) expression in EAE was suppressed by CYM-5442 treatment, relative to vehicle, and levels of key cytokines, such as interleukin 17A, were also significantly reduced in drug-treated mice. S1P(1)-selective agonists that induce reversible lymphopenia while persisting in the CNS may be effective MS treatments.


Asunto(s)
Linfopenia/tratamiento farmacológico , Esclerosis Múltiple/tratamiento farmacológico , Receptores de Lisoesfingolípidos/agonistas , Animales , Sistema Nervioso Central/metabolismo , Citocinas/efectos de los fármacos , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Clorhidrato de Fingolimod , Inmunosupresores , Indanos , Ratones , Oxadiazoles , Glicoles de Propileno/uso terapéutico , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/uso terapéutico
10.
Sci Adv ; 7(28)2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34233878

RESUMEN

T lymphocytes encounter complex mechanical cues during an immune response. The mechanosensitive ion channel, Piezo1, drives inflammatory responses to bacterial infections, wound healing, and cancer; however, its role in helper T cell function remains unclear. In an animal model for multiple sclerosis, experimental autoimmune encephalomyelitis (EAE), we found that mice with genetic deletion of Piezo1 in T cells showed diminished disease severity. Unexpectedly, Piezo1 was not essential for lymph node homing, interstitial motility, Ca2+ signaling, T cell proliferation, or differentiation into proinflammatory T helper 1 (TH1) and TH17 subsets. However, Piezo1 deletion in T cells resulted in enhanced transforming growth factor-ß (TGFß) signaling and an expanded pool of regulatory T (Treg) cells. Moreover, mice with deletion of Piezo1 specifically in Treg cells showed significant attenuation of EAE. Our results indicate that Piezo1 selectively restrains Treg cells, without influencing activation events or effector T cell functions.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Animales , Diferenciación Celular , Encefalomielitis Autoinmune Experimental/patología , Canales Iónicos/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Linfocitos T Reguladores , Células TH1
11.
Elife ; 72018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30382938

RESUMEN

Mechanically activated (MA) ion channels convert physical forces into electrical signals, and are essential for eukaryotic physiology. Despite their importance, few bona-fide MA channels have been described in plants and animals. Here, we show that various members of the OSCA and TMEM63 family of proteins from plants, flies, and mammals confer mechanosensitivity to naïve cells. We conclusively demonstrate that OSCA1.2, one of the Arabidopsis thaliana OSCA proteins, is an inherently mechanosensitive, pore-forming ion channel. Our results suggest that OSCA/TMEM63 proteins are the largest family of MA ion channels identified, and are conserved across eukaryotes. Our findings will enable studies to gain deep insight into molecular mechanisms of MA channel gating, and will facilitate a better understanding of mechanosensory processes in vivo across plants and animals.


Asunto(s)
Secuencia Conservada , Evolución Molecular , Activación del Canal Iónico , Canales Iónicos/genética , Canales Iónicos/metabolismo , Mecanotransducción Celular , Animales , Arabidopsis , Fenómenos Biofísicos , Gadolinio/farmacología , Células HEK293 , Humanos , Liposomas , Concentración Osmolar
12.
Elife ; 72018 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-30095067

RESUMEN

SWELL1 (LRRC8A) is the only essential subunit of the Volume Regulated Anion Channel (VRAC), which regulates cellular volume homeostasis and is activated by hypotonic solutions. SWELL1, together with four other LRRC8 family members, potentially forms a vastly heterogeneous cohort of VRAC channels with different properties; however, SWELL1 alone is also functional. Here, we report a high-resolution cryo-electron microscopy structure of full-length human homo-hexameric SWELL1. The structure reveals a trimer of dimers assembly with symmetry mismatch between the pore-forming domain and the cytosolic leucine-rich repeat (LRR) domains. Importantly, mutational analysis demonstrates that a charged residue at the narrowest constriction of the homomeric channel is an important pore determinant of heteromeric VRAC. Additionally, a mutation in the flexible N-terminal portion of SWELL1 affects pore properties, suggesting a putative link between intracellular structures and channel regulation. This structure provides a scaffold for further dissecting the heterogeneity and mechanism of activation of VRAC.


Asunto(s)
Proteínas de la Membrana/química , Multimerización de Proteína/genética , Relación Estructura-Actividad , Canales Aniónicos Dependientes del Voltaje/química , Aminoácidos/química , Aminoácidos/genética , Células HeLa , Humanos , Proteínas de la Membrana/genética , Familia de Multigenes , Mutación , Estructura Cuaternaria de Proteína , Canales Aniónicos Dependientes del Voltaje/genética
13.
Neuron ; 94(2): 266-270.e3, 2017 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-28426961

RESUMEN

A gold standard for characterizing mechanically activated (MA) currents is via heterologous expression of candidate channels in naive cells. Two recent studies described MA channels using this paradigm. TMEM150c was proposed to be a component of an MA channel partly based on a heterologous expression approach (Hong et al., 2016). In another study, Piezo1's N-terminal "propeller" domain was proposed to constitute an intrinsic mechanosensitive module based on expression of a chimera between a pore-forming domain of the mechanically insensitive ASIC1 channel and Piezo1 (Zhao et al., 2016). When we attempted to replicate these results, we found each construct conferred modest MA currents in a small fraction of naive HEK cells similar to the published work. Strikingly, these MA currents were not detected in cells in which endogenous Piezo1 was CRISPR/Cas9 inactivated. These results highlight the importance of choosing cells lacking endogenous MA channels to assay the mechanotransduction properties of various proteins. This Matters Arising paper is in response to Hong et al. (2016) and Zhao et al. (2016) in Neuron. See also the response papers by Hong et al. (2017) and Zhao et al. (2017) published concurrently with this Matters Arising.


Asunto(s)
Canales Iónicos/metabolismo , Mecanotransducción Celular/fisiología , Neuronas/metabolismo , Transporte Biológico , Línea Celular , Humanos , Mutagénesis Insercional/métodos
14.
Elife ; 42015 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-26001274

RESUMEN

Red blood cells (RBCs) experience significant mechanical forces while recirculating, but the consequences of these forces are not fully understood. Recent work has shown that gain-of-function mutations in mechanically activated Piezo1 cation channels are associated with the dehydrating RBC disease xerocytosis, implicating a role of mechanotransduction in RBC volume regulation. However, the mechanisms by which these mutations result in RBC dehydration are unknown. In this study, we show that RBCs exhibit robust calcium entry in response to mechanical stretch and that this entry is dependent on Piezo1 expression. Furthermore, RBCs from blood-cell-specific Piezo1 conditional knockout mice are overhydrated and exhibit increased fragility both in vitro and in vivo. Finally, we show that Yoda1, a chemical activator of Piezo1, causes calcium influx and subsequent dehydration of RBCs via downstream activation of the KCa3.1 Gardos channel, directly implicating Piezo1 signaling in RBC volume control. Therefore, mechanically activated Piezo1 plays an essential role in RBC volume homeostasis.


Asunto(s)
Calcio/metabolismo , Eritrocitos/fisiología , Canales Iónicos/fisiología , Mecanotransducción Celular/fisiología , Análisis de Varianza , Animales , Fenómenos Biomecánicos , Western Blotting , Cartilla de ADN/genética , Ensayo de Inmunoadsorción Enzimática , Recuento de Eritrocitos , Eritrocitos/metabolismo , Eritrocitos/ultraestructura , Citometría de Flujo , Fluorescencia , Canales Iónicos/genética , Canales Iónicos/metabolismo , Ratones , Ratones Noqueados , Microscopía Electrónica de Rastreo , Mutación/genética , Bibliotecas de Moléculas Pequeñas/farmacología
15.
Nat Commun ; 6: 8329, 2015 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-26387913

RESUMEN

Piezo1 ion channels are mediators of mechanotransduction in several cell types including the vascular endothelium, renal tubular cells and erythrocytes. Gain-of-function mutations in PIEZO1 cause an autosomal dominant haemolytic anaemia in humans called dehydrated hereditary stomatocytosis. However, the phenotypic consequence of PIEZO1 loss of function in humans has not previously been documented. Here we discover a novel role of this channel in the lymphatic system. Through whole-exome sequencing, we identify biallelic mutations in PIEZO1 (a splicing variant leading to early truncation and a non-synonymous missense variant) in a pair of siblings affected with persistent lymphoedema caused by congenital lymphatic dysplasia. Analysis of patients' erythrocytes as well as studies in a heterologous system reveal greatly attenuated PIEZO1 function in affected alleles. Our results delineate a novel clinical category of PIEZO1-associated hereditary lymphoedema.


Asunto(s)
Anemia Hemolítica Congénita/metabolismo , Hidropesía Fetal/metabolismo , Canales Iónicos/metabolismo , Enfermedades Linfáticas/metabolismo , Secuencia de Aminoácidos , Anemia Hemolítica Congénita/genética , Preescolar , Eritrocitos/metabolismo , Femenino , Genes Recesivos , Humanos , Hidropesía Fetal/genética , Lactante , Canales Iónicos/química , Canales Iónicos/genética , Enfermedades Linfáticas/genética , Masculino , Datos de Secuencia Molecular , Mutación , Mutación Missense , Alineación de Secuencia
17.
Science ; 335(6070): 851-5, 2012 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-22344443

RESUMEN

The lyso-phospholipid sphingosine 1-phosphate modulates lymphocyte trafficking, endothelial development and integrity, heart rate, and vascular tone and maturation by activating G protein-coupled sphingosine 1-phosphate receptors. Here, we present the crystal structure of the sphingosine 1-phosphate receptor 1 fused to T4-lysozyme (S1P(1)-T4L) in complex with an antagonist sphingolipid mimic. Extracellular access to the binding pocket is occluded by the amino terminus and extracellular loops of the receptor. Access is gained by ligands entering laterally between helices I and VII within the transmembrane region of the receptor. This structure, along with mutagenesis, agonist structure-activity relationship data, and modeling, provides a detailed view of the molecular recognition and requirement for hydrophobic volume that activates S1P(1), resulting in the modulation of immune and stromal cell responses.


Asunto(s)
Receptores de Lisoesfingolípidos/química , Anilidas/química , Sitios de Unión , Cristalografía por Rayos X , Modelos Moleculares , Muramidasa/química , Mutagénesis , Organofosfonatos/química , Conformación Proteica , Receptores de Lisoesfingolípidos/agonistas , Receptores de Lisoesfingolípidos/antagonistas & inhibidores , Receptores de Lisoesfingolípidos/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética
18.
Trends Immunol ; 28(3): 102-7, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17276731

RESUMEN

The lysophospholipid sphingosine 1-phosphate (S1P) is a pleiotropic signaling lipid present constitutively in plasma, and secreted locally at elevated concentrations at sites of inflammation. S1P maintains essential variable homeostatic functions in addition to inducing pathophysiology through the activation of five specific high-affinity G-protein-coupled receptors. Therefore, S1P can function as an extracellular rheostat regulating tonic and acutely evoked functions. Although S1P receptors can regulate lymphoid development and lymphocyte trafficking, and different opinions exist on the roles of receptor agonism and functional antagonism in regulating lymphocyte recirculation, this personal perspective highlights the pivotal control points regulated by constitutive and induced S1P receptor tone at vascular endothelial and lymphatic endothelial barriers, through which S1P agonism impacts on both innate and adaptive immunity. We also emphasize how specific, proof-of-concept chemical tools complement genetic approaches by enabling reversible perturbation of the S1P-S1P(1) receptor axis and, thus, clarifying in vivo mechanisms in the absence of developmental compensations.


Asunto(s)
Endotelio Vascular/metabolismo , Lisofosfolípidos/fisiología , Esfingosina/análogos & derivados , Animales , Antígenos CD/fisiología , Antígenos de Diferenciación de Linfocitos T/fisiología , Movimiento Celular , Clorhidrato de Fingolimod , Humanos , Lectinas Tipo C , Linfocitos/fisiología , Glicoles de Propileno/farmacología , Receptores de Lisoesfingolípidos/fisiología , Transducción de Señal , Esfingosina/farmacología , Esfingosina/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA