Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 44(4): 782-94, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27037192

RESUMEN

Activation of T cells is mediated by the engagement of T cell receptors (TCRs) followed by calcium entry via store-operated calcium channels. Here we have shown an additional route for calcium entry into T cells-through the low-voltage-activated T-type CaV3.1 calcium channel. CaV3.1 mediated a substantial current at resting membrane potentials, and its deficiency had no effect on TCR-initiated calcium entry. Mice deficient for CaV3.1 were resistant to the induction of experimental autoimmune encephalomyelitis and had reduced productions of the granulocyte-macrophage colony-stimulating factor (GM-CSF) by central nervous system (CNS)-infiltrating T helper 1 (Th1) and Th17 cells. CaV3.1 deficiency led to decreased secretion of GM-CSF from in vitro polarized Th1 and Th17 cells. Nuclear translocation of the nuclear factor of activated T cell (NFAT) was also reduced in CaV3.1-deficient T cells. These data provide evidence for T-type channels in immune cells and their potential role in shaping the autoimmune response.


Asunto(s)
Canales de Calcio Tipo T/genética , Encefalomielitis Autoinmune Experimental/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Factores de Transcripción NFATC/metabolismo , Células TH1/inmunología , Células Th17/inmunología , Transporte Activo de Núcleo Celular/genética , Animales , Autoinmunidad/genética , Autoinmunidad/inmunología , Calcio/metabolismo , Citocinas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
2.
Proc Natl Acad Sci U S A ; 119(1)2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34949717

RESUMEN

Airway remodeling and airway hyperresponsiveness are central drivers of asthma severity. Airway remodeling is a structural change involving the dedifferentiation of airway smooth muscle (ASM) cells from a quiescent to a proliferative and secretory phenotype. Here, we show up-regulation of the endoplasmic reticulum Ca2+ sensor stromal-interacting molecule 1 (STIM1) in ASM of asthmatic mice. STIM1 is required for metabolic and transcriptional reprogramming that supports airway remodeling, including ASM proliferation, migration, secretion of cytokines and extracellular matrix, enhanced mitochondrial mass, and increased oxidative phosphorylation and glycolytic flux. Mechanistically, STIM1-mediated Ca2+ influx is critical for the activation of nuclear factor of activated T cells 4 and subsequent interleukin-6 secretion and transcription of pro-remodeling transcription factors, growth factors, surface receptors, and asthma-associated proteins. STIM1 drives airway hyperresponsiveness in asthmatic mice through enhanced frequency and amplitude of ASM cytosolic Ca2+ oscillations. Our data advocates for ASM STIM1 as a target for asthma therapy.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Asma/fisiopatología , Músculo Liso/fisiopatología , Hipersensibilidad Respiratoria , Molécula de Interacción Estromal 1/fisiología , Animales , Asma/patología , Calcio/metabolismo , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Reprogramación Celular/fisiología , Enfermedad Crónica , Transporte Iónico , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Músculo Liso/patología , Molécula de Interacción Estromal 1/genética , Transcripción Genética/fisiología
3.
J Cell Sci ; 134(21)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34622926

RESUMEN

Protein kinase C (PKC)-ε is required for membrane addition during IgG-mediated phagocytosis, but its role in this process is ill defined. Here, we performed high-resolution imaging, which reveals that PKC-ε exits the Golgi and enters phagosomes on vesicles that then fuse. TNF and PKC-ε colocalize at the Golgi and on vesicles that enter the phagosome. Loss of PKC-ε and TNF delivery upon nocodazole treatment confirmed vesicular transport on microtubules. That TNF+ vesicles were not delivered in macrophages from PKC-ε null mice, or upon dissociation of the Golgi-associated pool of PKC-ε, implies that Golgi-tethered PKC-ε is a driver of Golgi-to-phagosome trafficking. Finally, we established that the regulatory domain of PKC-ε is sufficient for delivery of TNF+ vesicles to the phagosome. These studies reveal a novel role for PKC-ε in focal exocytosis - its regulatory domain drives Golgi-derived vesicles to the phagosome, whereas catalytic activity is required for their fusion. This is one of the first examples of a PKC requirement for vesicular trafficking and describes a novel function for a PKC regulatory domain. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Fagocitosis , Proteína Quinasa C-epsilon , Animales , Exocitosis , Inmunoglobulina G , Ratones , Fagosomas
4.
Cell Mol Life Sci ; 79(6): 284, 2022 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-35526196

RESUMEN

BACKGROUND AND AIMS: Recent evidences highlight a role of the mitochondria calcium homeostasis in the development of colorectal cancer (CRC). To overcome treatment resistance, we aimed to evaluate the role of the mitochondrial sodium-calcium-lithium exchanger (NCLX) and its targeting in CRC. We also identified curcumin as a new inhibitor of NCLX. METHODS: We examined whether curcumin and pharmacological compounds induced the inhibition of NCLX-mediated mitochondrial calcium (mtCa2+) extrusion, the role of redox metabolism in this process. We evaluated their anti-tumorigenic activity in vitro and in a xenograft mouse model. We analyzed NCLX expression and associations with survival in The Cancer Genome Atlas (TCGA) dataset and in tissue microarrays from 381 patients with microsatellite instability (MSI)-driven CRC. RESULTS: In vitro, curcumin exerted strong anti-tumoral activity through its action on NCLX with mtCa2+ and reactive oxygen species overload associated with a mitochondrial membrane depolarization, leading to reduced ATP production and apoptosis. NCLX inhibition with pharmacological and molecular approaches reproduced the effects of curcumin. NCLX inhibitors decreased CRC tumor growth in vivo. Both transcriptomic analysis of TCGA dataset and immunohistochemical analysis of tissue microarrays demonstrated that higher NCLX expression was associated with MSI status, and for the first time, NCLX expression was significantly associated with recurrence-free survival. CONCLUSIONS: Our findings highlight a novel anti-tumoral mechanism of curcumin through its action on NCLX and mitochondria calcium overload that could benefit for therapeutic schedule of patients with MSI CRC.


Asunto(s)
Neoplasias Colorrectales , Curcumina , Inestabilidad de Microsatélites , Intercambiador de Sodio-Calcio , Animales , Calcio/metabolismo , Señalización del Calcio , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Curcumina/farmacología , Humanos , Ratones , Repeticiones de Microsatélite , Proteínas Mitocondriales/metabolismo , Intercambiador de Sodio-Calcio/antagonistas & inhibidores
5.
Proc Natl Acad Sci U S A ; 117(29): 17369-17380, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32641503

RESUMEN

Voltage-gated L-type Ca2+ channel (Cav1.2) blockers (LCCBs) are major drugs for treating hypertension, the preeminent risk factor for heart failure. Vascular smooth muscle cell (VSMC) remodeling is a pathological hallmark of chronic hypertension. VSMC remodeling is characterized by molecular rewiring of the cellular Ca2+ signaling machinery, including down-regulation of Cav1.2 channels and up-regulation of the endoplasmic reticulum (ER) stromal-interacting molecule (STIM) Ca2+ sensor proteins and the plasma membrane ORAI Ca2+ channels. STIM/ORAI proteins mediate store-operated Ca2+ entry (SOCE) and drive fibro-proliferative gene programs during cardiovascular remodeling. SOCE is activated by agonists that induce depletion of ER Ca2+, causing STIM to activate ORAI. Here, we show that the three major classes of LCCBs activate STIM/ORAI-mediated Ca2+ entry in VSMCs. LCCBs act on the STIM N terminus to cause STIM relocalization to junctions and subsequent ORAI activation in a Cav1.2-independent and store depletion-independent manner. LCCB-induced promotion of VSMC remodeling requires STIM1, which is up-regulated in VSMCs from hypertensive rats. Epidemiology showed that LCCBs are more associated with heart failure than other antihypertensive drugs in patients. Our findings unravel a mechanism of LCCBs action on Ca2+ signaling and demonstrate that LCCBs promote vascular remodeling through STIM-mediated activation of ORAI. Our data indicate caution against the use of LCCBs in elderly patients or patients with advanced hypertension and/or onset of cardiovascular remodeling, where levels of STIM and ORAI are elevated.


Asunto(s)
Canales de Calcio Tipo L/metabolismo , Hipertensión/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Molécula de Interacción Estromal 2/metabolismo , Moléculas de Interacción Estromal/metabolismo , Remodelación Vascular/fisiología , Animales , Antihipertensivos/farmacología , Calcio/metabolismo , Canales de Calcio Tipo L/efectos de los fármacos , Membrana Celular/metabolismo , Movimiento Celular , Proliferación Celular , Modelos Animales de Enfermedad , Retículo Endoplásmico/metabolismo , Técnicas de Inactivación de Genes , Células HEK293 , Insuficiencia Cardíaca , Humanos , Proteínas de la Membrana/genética , Miocitos del Músculo Liso , Proteínas de Neoplasias , Proteína ORAI1/genética , Ratas , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 2/genética
6.
Angew Chem Int Ed Engl ; 62(9): e202217724, 2023 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-36625565

RESUMEN

We report the first highly enantioselective construction of silicon-stereocenters by asymmetric enamine catalysis. An unprecedented desymmetric intramolecular aldolization of prochiral siladials was thus developed for the facile access of multifunctional silicon-stereogenic silacycles in high to excellent enantioselectivity. With an enal moiety, these adducts could be readily elaborated for the diverse synthesis of silicon-stereogenic compounds, and for late-stage modification.

7.
J Biol Chem ; 297(4): 101174, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34499925

RESUMEN

Mitochondrial Ca2+ uptake tailors the strength of stimulation of plasma membrane phospholipase C-coupled receptors to that of cellular bioenergetics. However, how Ca2+ uptake by the mitochondrial Ca2+ uniporter (MCU) shapes receptor-evoked interorganellar Ca2+ signaling is unknown. Here, we used CRISPR/Cas9 gene knockout, subcellular Ca2+ imaging, and mathematical modeling to show that MCU is a universal regulator of intracellular Ca2+ signaling across mammalian cell types. MCU activity sustains cytosolic Ca2+ signaling by preventing Ca2+-dependent inactivation of store-operated Ca2+ release-activated Ca2+ channels and by inhibiting Ca2+ extrusion. Paradoxically, MCU knockout (MCU-KO) enhanced cytosolic Ca2+ responses to store depletion. Physiological agonist stimulation in MCU-KO cells led to enhanced frequency of cytosolic Ca2+ oscillations, endoplasmic reticulum Ca2+ refilling, nuclear translocation of nuclear factor for activated T cells transcription factors, and cell proliferation, without altering inositol-1,4,5-trisphosphate receptor activity. Our data show that MCU has dual counterbalancing functions at the cytosol-mitochondria interface, whereby the cell-specific MCU-dependent cytosolic Ca2+ clearance and buffering capacity of mitochondria reciprocally regulate interorganellar Ca2+ transfer and nuclear factor for activated T cells nuclear translocation during receptor-evoked signaling. These findings highlight the critical dual function of the MCU not only in the acute Ca2+ buffering by mitochondria but also in shaping endoplasmic reticulum and cytosolic Ca2+ signals that regulate cellular transcription and function.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio , Calcio/metabolismo , Citosol/metabolismo , Mitocondrias/metabolismo , Factores de Transcripción NFATC/metabolismo , Sistemas CRISPR-Cas , Canales de Calcio/genética , Retículo Endoplásmico , Técnicas de Inactivación de Genes , Células HCT116 , Células HEK293 , Humanos , Células Jurkat , Activación de Linfocitos , Factores de Transcripción NFATC/genética , Linfocitos T/metabolismo
8.
EMBO J ; 36(6): 797-815, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28219928

RESUMEN

Mitochondria exert important control over plasma membrane (PM) Orai1 channels mediating store-operated Ca2+ entry (SOCE). Although the sensing of endoplasmic reticulum (ER) Ca2+ stores by STIM proteins and coupling to Orai1 channels is well understood, how mitochondria communicate with Orai1 channels to regulate SOCE activation remains elusive. Here, we reveal that SOCE is accompanied by a rise in cytosolic Na+ that is critical in activating the mitochondrial Na+/Ca2+ exchanger (NCLX) causing enhanced mitochondrial Na+ uptake and Ca2+ efflux. Omission of extracellular Na+ prevents the cytosolic Na+ rise, inhibits NCLX activity, and impairs SOCE and Orai1 channel current. We show further that SOCE activates a mitochondrial redox transient which is dependent on NCLX and is required for preventing Orai1 inactivation through oxidation of a critical cysteine (Cys195) in the third transmembrane helix of Orai1. We show that mitochondrial targeting of catalase is sufficient to rescue redox transients, SOCE, and Orai1 currents in NCLX-deficient cells. Our findings identify a hitherto unknown NCLX-mediated pathway that coordinates Na+ and Ca2+ signals to effect mitochondrial redox control over SOCE.


Asunto(s)
Calcio/metabolismo , Mitocondrias/metabolismo , Proteína ORAI1/metabolismo , Intercambiador de Sodio-Calcio/metabolismo , Sodio/metabolismo , Línea Celular , Humanos , Proteínas Mitocondriales , Oxidación-Reducción
9.
FASEB J ; 34(5): 6871-6887, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32248623

RESUMEN

This study used constitutive CD226 gene knockout (KO) mice as a model to investigate the functions and mechanisms of CD226 in megakaryocyte (MK) maturation and platelet activation. Although CD226 deficiency did not cause MK polyploidization or platelet granule abnormalities, increased MK counts were detected in the femora bone marrow (BM) and spleen of CD226 KO mice. Particularly, CD226 KO mice have a more extensive membrane system in MKs and platelets than wild-type (WT) mice. We also demonstrated that CD226 KO mice displayed increased platelet counts, shortened bleeding time, and enhanced platelet aggregation. CD226 KO platelets had an increased mature platelet ratio compared to the control platelets. In addition, the observed reduction in bleeding time may be due to decreased nitric oxide (NO) production in the platelets. Platelet-specific CD226-deficient mice showed similar increased MK counts, shortened bleeding time, enhanced platelet aggregation, and decreased NO production in platelets. Furthermore, we performed middle cerebral artery occlusion-reperfusion surgery on WT and CD226 KO mice to explore the potential effect of CD226 on acute ischemia-reperfusion injury; the results revealed that CD226 deficiency led to significantly increased infarct area. Thus, CD226 is a promising candidate for the treatment of thrombotic disorders.


Asunto(s)
Antígenos de Diferenciación de Linfocitos T/sangre , Megacariocitos/citología , Megacariocitos/fisiología , Activación Plaquetaria/fisiología , Animales , Antígenos de Diferenciación de Linfocitos T/genética , Plaquetas/fisiología , Plaquetas/ultraestructura , Isquemia Encefálica/sangre , Isquemia Encefálica/genética , Isquemia Encefálica/patología , Modelos Animales de Enfermedad , Femenino , Integrina beta3/sangre , Masculino , Megacariocitos/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Activación Plaquetaria/genética , Adhesividad Plaquetaria/genética , Adhesividad Plaquetaria/fisiología , Agregación Plaquetaria/genética , Agregación Plaquetaria/fisiología , Recuento de Plaquetas , Trombopoyesis/genética , Trombopoyesis/fisiología
10.
J Biol Chem ; 294(16): 6318-6332, 2019 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-30824535

RESUMEN

Store-operated Ca2+ entry (SOCE) is a ubiquitous pathway for Ca2+ influx across the plasma membrane (PM). SOCE is mediated by the endoplasmic reticulum (ER)-associated Ca2+-sensing proteins stromal interaction molecule 1 (STIM1) and STIM2, which transition into an active conformation in response to ER Ca2+ store depletion, thereby interacting with and gating PM-associated ORAI1 channels. Although structurally homologous, STIM1 and STIM2 generate distinct Ca2+ signatures in response to varying strengths of agonist stimulation. The physiological functions of these Ca2+ signatures, particularly under native conditions, remain unclear. To investigate the structural properties distinguishing STIM1 and STIM2 activation of ORAI1 channels under native conditions, here we used CRISPR/Cas9 to generate STIM1-/-, STIM2-/-, and STIM1/2-/- knockouts in HEK293 and colorectal HCT116 cells. We show that depending on cell type, STIM2 can significantly sustain SOCE in response to maximal store depletion. Utilizing the SOCE modifier 2-aminoethoxydiphenyl borate (2-APB), we demonstrate that 2-APB-activated store-independent Ca2+ entry is mediated exclusively by endogenous STIM2. Using variants that either stabilize or disrupt intramolecular interactions of STIM C termini, we show that the increased flexibility of the STIM2 C terminus contributes to its selective store-independent activation by 2-APB. However, STIM1 variants with enhanced flexibility in the C terminus failed to support its store-independent activation. STIM1/STIM2 chimeric constructs indicated that coordination between N-terminal sensitivity and C-terminal flexibility is required for specific store-independent STIM2 activation. Our results clarify the structural determinants underlying activation of specific STIM isoforms, insights that are potentially useful for isoform-selective drug targeting.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Molécula de Interacción Estromal 2/metabolismo , Compuestos de Boro/química , Compuestos de Boro/farmacología , Calcio/química , Retículo Endoplásmico/química , Retículo Endoplásmico/genética , Técnicas de Silenciamiento del Gen , Células HCT116 , Células HEK293 , Humanos , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Dominios Proteicos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Molécula de Interacción Estromal 1/química , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo , Molécula de Interacción Estromal 2/química , Molécula de Interacción Estromal 2/genética
11.
Angew Chem Int Ed Engl ; 59(4): 1634-1643, 2020 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-31755631

RESUMEN

Azacycles such as indoles and tetrahydroquinolines are privileged structures in drug development. Reported here is an unprecedented regiodivergent intramolecular nucleophilic addition reaction of imines as a flexible approach to access N-functionalized indoles and tetrahydroquinolines, by the control of reaction at the N-terminus and C-terminus, respectively. Using ketimines derived from 2-(2-nitroethyl)anilines with isatins or α-ketoesters, the regioselective N-attack reaction gives N-functionalized indoles, while the catalytic enantioselective C-attack reaction affords chiral tetrahydroquinolines featuring an α-tetrasubstituted stereocenter. Mechanistic studies reveal that hydrogen-bonding interactions may greatly facilitate such unusual N-attack reactions of imines. The utility of this protocol is highlighted by the catalytic enantioselective formal synthesis of (-)-psychotrimine, and the construction of various fused aza-heterocycles.

12.
Org Biomol Chem ; 17(8): 2174-2181, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30720040

RESUMEN

An efficient Pd/Zn co-catalyzed chemo-selective hydrogenation of α-methylene-γ-keto carboxylic acids is described. This methodology offers a divergent synthesis of α-methyl-γ-keto carboxylic acids, α-methylcarboxylic acids, and lactones starting from α-methylene-γ-keto carboxylic acids via selective hydrogenation by varying the catalytic conditions avoiding the use of high pressure of hydrogen. The reaction also showed broad substrate tolerance to give the desired products in good to excellent yields.

13.
J Immunol ; 199(1): 271-277, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28539432

RESUMEN

Protein kinase C-ε (PKC-ε) at phagocytic cups mediates the membrane fusion necessary for efficient IgG-mediated phagocytosis. The C1B and pseudosubstrate (εPS) domains are necessary and sufficient for this concentration. C1B binds diacylglycerol; the docking partner for εPS is unknown. Liposome assays revealed that the εPS binds phosphatidylinositol 4-phosphate (PI4P) and PI(3,5)P2 Wortmannin, but not LY294002, inhibits PKC-ε concentration at cups and significantly reduces the rate of phagocytosis. As Wortmannin inhibits PI4 kinase, we hypothesized that PI4P mediates the PKC-ε concentration at cups and the rate of phagocytosis. PKC-ε colocalizes with the trans-Golgi network (TGN) PI4P reporter, P4M, suggesting it is tethered at the TGN. Real-time imaging of GFP-PKC-ε-expressing macrophages revealed a loss of Golgi-associated PKC-ε during phagocytosis, consistent with a Golgi-to-phagosome translocation. Treatment with PIK93, a PI4 kinase inhibitor, reduces PKC-ε at both the TGN and the cup, decreases phagocytosis, and prevents the increase in capacitance that accompanies membrane fusion. Finally, expression of the Golgi-directed PI4P phosphatase, hSac1-K2A, recapitulates the PIK93 phenotype, confirming that Golgi-associated PI4P is critical for efficient phagocytosis. Together these data are consistent with a model in which PKC-ε is tethered to the TGN via an εPS-PI4P interaction. The TGN-associated pool of PKC-ε concentrates at the phagocytic cup where it mediates the membrane fusion necessary for phagocytosis. The novelty of these data lies in the demonstration that εPS binds PI4P and PI(3,5)P2 and that PI4P is necessary for PKC-ε localization at the TGN, its translocation to the phagocytic cup, and the membrane fusion required for efficient Fc [γ] receptor-mediated phagocytosis.


Asunto(s)
Fagocitosis , Fagosomas/inmunología , Fosfatos de Fosfatidilinositol/metabolismo , Proteína Quinasa C-epsilon/metabolismo , Animales , Membrana Celular/metabolismo , Aparato de Golgi/metabolismo , Proteínas Fluorescentes Verdes , Fusión de Membrana , Ratones , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Fagosomas/metabolismo , Proteína Quinasa C/inmunología , Proteína Quinasa C/metabolismo , Transducción de Señal
14.
Physiol Plant ; 164(3): 242-250, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29235671

RESUMEN

Forward genetic analysis, widely used to find new gene functions, benefits from the availability of mutants. At present, based on Agrobacterium-mediated plant transformation technology, many transfer (T)-DNA transformants have been created. However, cloning their T-DNA insertion sites, which enables identification of the mutated genes, is still challenging. In this study, we improved adapter ligation-mediated polymerase chain reaction (A-PCR), which mainly utilizes the Thermal Asymmetric interlaced reaction and Degenerate sequence-recognizing restriction Endonucleases (TADE). Using the new method TADE-mediated A-PCR (TADEA-PCR), we successfully cloned 22 of all the 24 junction sites in 10 Arabidopsis thaliana L. transformants that contained 12 T-DNA insertions in total, giving a success rate of 91.7%. In most cases, the two junction sites resulting from a single T-DNA insertion were simultaneously cloned. In addition, TADEA-PCR was able to clone more than two junction sites present in one transformant containing several T-DNA insertions. Overall, TADEA-PCR is a powerful technique for cloning T-DNA insertion sites.


Asunto(s)
ADN Bacteriano/genética , Reacción en Cadena de la Polimerasa/métodos , Mutación , Análisis de Secuencia de ADN
15.
J Biol Chem ; 291(44): 22894-22912, 2016 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-27624938

RESUMEN

Endothelial barrier function is tightly regulated by plasma membrane receptors and is crucial for tissue fluid homeostasis; its dysfunction causes disease, including sepsis and inflammation. The ubiquitous activation of Ca2+ signaling upon phospholipase C-coupled receptor ligation leads quite naturally to the assumption that Ca2+ signaling is required for receptor-regulated endothelial barrier function. This widespread hypothesis draws analogy from smooth muscle and proposes the requirement of G protein-coupled receptor (GPCR)-generated Ca2+ signaling in activating the endothelial contractile apparatus and generating interendothelial gaps. Notwithstanding endothelia being non-excitable in nature, the hypothesis of Ca2+-induced endothelial contraction has been invoked to explain actions of GPCR agonists that either disrupt or stabilize endothelial barrier function. Here, we challenge this correlative hypothesis by showing a lack of causal link between GPCR-generated Ca2+ signaling and changes in human microvascular endothelial barrier function. We used three endogenous GPCR agonists: thrombin and histamine, which disrupt endothelial barrier function, and sphingosine-1-phosphate, which stabilizes barrier function. The qualitatively different effects of these three agonists on endothelial barrier function occur independently of Ca2+ entry through the ubiquitous store-operated Ca2+ entry channel Orai1, global Ca2+ entry across the plasma membrane, and Ca2+ release from internal stores. However, disruption of endothelial barrier function by thrombin and histamine requires the Ca2+ sensor stromal interacting molecule-1 (STIM1), whereas sphingosine-1-phosphate-mediated enhancement of endothelial barrier function occurs independently of STIM1. We conclude that although STIM1 is required for GPCR-mediated disruption of barrier function, a causal link between GPCR-induced cytoplasmic Ca2+ increases and acute changes in barrier function is missing. Thus, the cytosolic Ca2+-induced endothelial contraction is a cum hoc fallacy that should be abandoned.


Asunto(s)
Señalización del Calcio , Células Endoteliales/metabolismo , Calcio/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Lisofosfolípidos/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo , Trombina/genética , Trombina/metabolismo
16.
J Biol Chem ; 290(8): 5015-5027, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25540197

RESUMEN

Leukotriene-C4 synthase (LTC4S) generates LTC4 from arachidonic acid metabolism. LTC4 is a proinflammatory factor that acts on plasma membrane cysteinyl leukotriene receptors. Recently, however, we showed that LTC4 was also a cytosolic second messenger that activated store-independent LTC4-regulated Ca(2+) (LRC) channels encoded by Orai1/Orai3 heteromultimers in vascular smooth muscle cells (VSMCs). We showed that Orai3 and LRC currents were up-regulated in medial and neointimal VSMCs after vascular injury and that Orai3 knockdown inhibited LRC currents and neointimal hyperplasia. However, the role of LTC4S in neointima formation remains unknown. Here we show that LTC4S knockdown inhibited LRC currents in VSMCs. We performed in vivo experiments where rat left carotid arteries were injured using balloon angioplasty to cause neointimal hyperplasia. Neointima formation was associated with up-regulation of LTC4S protein expression in VSMCs. Inhibition of LTC4S expression in injured carotids by lentiviral particles encoding shRNA inhibited neointima formation and inward and outward vessel remodeling. LRC current activation did not cause nuclear factor for activated T cells (NFAT) nuclear translocation in VSMCs. Surprisingly, knockdown of either LTC4S or Orai3 yielded more robust and sustained Akt1 and Akt2 phosphorylation on Ser-473/Ser-474 upon serum stimulation. LTC4S and Orai3 knockdown inhibited VSMC migration in vitro with no effect on proliferation. Akt activity was suppressed in neointimal and medial VSMCs from injured vessels at 2 weeks postinjury but was restored when the up-regulation of either LTC4S or Orai3 was prevented by shRNA. We conclude that LTC4S and Orai3 altered Akt signaling to promote VSMC migration and neointima formation.


Asunto(s)
Canales de Calcio/metabolismo , Glutatión Transferasa/biosíntesis , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Neointima/metabolismo , Animales , Canales de Calcio/genética , Regulación Enzimológica de la Expresión Génica/genética , Glutatión Transferasa/genética , Hiperplasia/genética , Hiperplasia/metabolismo , Hiperplasia/patología , Masculino , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Neointima/genética , Neointima/patología , Proteína ORAI1 , Fosforilación/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/genética
17.
Adv Exp Med Biol ; 898: 251-64, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27161232

RESUMEN

Transient receptor potential canonical subfamily, member 7 (TRPC7) is the most recently identified member of the TRPC family of Ca(2+)-permeable non-selective cation channels. The gene encoding the TRPC7 channel plasma membrane protein was first cloned from mouse brain. TRPC7 mRNA and protein have been detected in cell types derived from multiple organ systems from various species including humans. Gq-coupled protein receptor activation is the predominant mode of TRPC7 activation. Lipid metabolites involved in the phospholipase C (PLC) signaling pathway, including diacylglycerol (DAG) and its precursor the phosphatidylinositol-4,5-bisphosphate (PIP2), have been shown to be direct regulators of TRPC7 channel. TRPC7 channels have been linked to the regulation of various cellular functions however, the depth of our understanding of TRPC7 channel function and regulation is limited in comparison to other TRP channel family members. This review takes a historical look at our current knowledge of TRPC7 mechanisms of activation and its role in cellular physiology and pathophysiology.


Asunto(s)
Calcio/metabolismo , Canales Catiónicos TRPC/fisiología , Animales , Señalización del Calcio , Transporte Iónico , Ratones , Conformación Proteica , Canales Catiónicos TRPC/química , Canales Catiónicos TRPC/metabolismo
18.
Circ Res ; 112(7): 1013-25, 2013 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-23349245

RESUMEN

RATIONALE: Through largely unknown mechanisms, Ca(2+) signaling plays important roles in vascular smooth muscle cell (VSMC) remodeling. Orai1-encoded store-operated Ca(2+) entry has recently emerged as an important player in VSMC remodeling. However, the role of the exclusively mammalian Orai3 protein in native VSMC Ca(2+) entry pathways, its upregulation during VSMC remodeling, and its contribution to neointima formation remain unknown. OBJECTIVE: The goal of this study was to determine the agonist-evoked Ca(2+) entry pathway contributed by Orai3; Orai3 potential upregulation and role during neointima formation after balloon injury of rat carotid arteries. METHODS AND RESULTS: Ca(2+) imaging and patch-clamp recordings showed that although the platelet-derived growth factor activates the canonical Ca(2+) release-activated Ca(2+) channels via store depletion in VSMC, the pathophysiological agonist thrombin activates a distinct Ca(2+)-selective channel contributed by Orai1, Orai3, and stromal interacting molecule1 in the same cells. Unexpectedly, Ca(2+) store depletion is not required for activation of Orai1/3 channel by thrombin. Rather, the signal for Orai1/3 channel activation is cytosolic leukotrieneC4 produced downstream thrombin receptor stimulation through the catalytic activity of leukotrieneC4 synthase. Importantly, Orai3 is upregulated in an animal model of VSMC neointimal remodeling, and in vivo Orai3 knockdown inhibits neointima formation. CONCLUSIONS: These results demonstrate that distinct native Ca(2+)-selective Orai channels are activated by different agonists/pathways and uncover a mechanism whereby leukotrieneC4 acts through hitherto unknown intracrine mode to elicit store-independent Ca(2+) signaling that promotes vascular occlusive disease. Orai3 and Orai3-containing channels provide novel targets for control of VSMC remodeling during vascular injury or disease.


Asunto(s)
Canales de Calcio/fisiología , Traumatismos de las Arterias Carótidas/fisiopatología , Leucotrieno C4/metabolismo , Músculo Liso Vascular/fisiopatología , Neointima/fisiopatología , Angioplastia de Balón/efectos adversos , Animales , Canales de Calcio/genética , Canales de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/fisiología , Traumatismos de las Arterias Carótidas/etiología , Traumatismos de las Arterias Carótidas/patología , Citosol/metabolismo , Modelos Animales de Enfermedad , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Músculo Liso Vascular/patología , Neointima/etiología , Neointima/patología , Proteína ORAI1 , Técnicas de Placa-Clamp , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Molécula de Interacción Estromal 1 , Trombina/metabolismo , Trombina/farmacología
19.
FASEB J ; 27(1): 63-75, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22993197

RESUMEN

Store-operated Ca(2+) entry (SOCE) encoded by Orai1 proteins is a ubiquitous Ca(2+)-selective conductance involved in cellular proliferation and migration. We recently described up-regulation of Orai3 channels that selectively mediate SOCE in estrogen receptor α-expressing (ERα(+)) breast cancer cells. However, the connection between ERα and Orai3 and the role of Orai3 in tumorigenesis remain unknown. Here, we show that ERα knockdown decreases Orai3 mRNA (by ∼63%) and protein (by ∼44%) with no effect on Orai1. ERα knockdown decreases Orai3-mediated SOCE (by ∼43%) and the corresponding Ca(2+) release-activated Ca(2+) (CRAC) current (by ∼42%) in ERα(+) MCF7 cells. The abrogation of SOCE in MCF7 cells on ERα knockdown can be rescued by ectopic expression of Orai3. ERα activation increased Orai3 expression and SOCE in MCF7 cells. Epidermal growth factor (EGF) and thrombin stimulate Ca(2+) influx into MCF7 cells through Orai3. Orai3 knockdown inhibited SOCE-dependent phosphorylation of extracellular signal-regulated kinase (ERK1/2; by ∼44%) and focal adhesion kinase (FAK; by ∼46%) as well as transcriptional activity of nuclear factor for activated T cells (NFAT; by ∼49%). Significantly, Orai3 knockdown selectively decreased anchorage-independent growth (by ∼58%) and Matrigel invasion (by ∼44%) of ERα(+) MCF7 cells with no effect on ERα(-) MDA-MB231 cells. Moreover, Orai3 knockdown inhibited ERα(+) cell tumorigenesis in immunodeficient mice (∼66% reduction in tumor volume). These data establish Orai3 as an ERα-regulated channel and a potential selective therapeutic target for ERα(+) breast cancers.


Asunto(s)
Canales de Calcio/fisiología , Transformación Celular Neoplásica , Receptor alfa de Estrógeno/fisiología , Animales , Western Blotting , Femenino , Humanos , Células MCF-7 , Ratones , Ratones SCID , Fosforilación , Reacción en Cadena de la Polimerasa
20.
Handb Exp Pharmacol ; 222: 189-204, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24756707

RESUMEN

Transient receptor potential canonical 7 (TRPC7) channel is the seventh member of the mammalian TRPC channel family. TRPC7 mRNA, protein, and channel activity have been detected in many tissues and organs from the mouse, rat, and human. TRPC7 has high sequence homology with TRPC3 and TRPC6, and all three channels are activated by membrane receptors that couple to isoforms of phospholipase C (PLC) and mediate non-selective cation currents. TRPC7, along with TRPC3 and TRPC6, can be activated by direct exogenous application of diacylglycerol (DAG) analogues and by pharmacological maneuvers that increase endogenous DAG in cells. TRPC7 shows distinct properties of activation, such as constitutive activity and susceptibility to negative regulation by extracellular Ca(2+) and by protein kinase C. TRPC7 can form heteromultimers with TRPC3 and TRPC6. Although TRPC7 remains one of the least studied TRPC channel, its role in various cell types and physiological and pathophysiological conditions is beginning to emerge.


Asunto(s)
Diglicéridos/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Permeabilidad de la Membrana Celular , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Activación del Canal Iónico , Potenciales de la Membrana , Ratones , Ratones Noqueados , Fenotipo , Conformación Proteica , Transducción de Señal , Relación Estructura-Actividad , Canales Catiónicos TRPC/química , Canales Catiónicos TRPC/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA