Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Proc Natl Acad Sci U S A ; 117(46): 29155-29165, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33139539

RESUMEN

LRRC8 family proteins on the plasma membrane play a critical role in cellular osmoregulation by forming volume-regulated anion channels (VRACs) necessary to prevent necrotic cell death. We demonstrate that intracellular LRRC8 proteins acting within lysosomes also play an essential role in cellular osmoregulation. LRRC8 proteins on lysosome membranes generate large lysosomal volume-regulated anion channel (Lyso-VRAC) currents in response to low cytoplasmic ionic strength conditions. When a double-leucine L706L707 motif at the C terminus of LRRC8A was mutated to alanines, normal plasma membrane VRAC currents were still observed, but Lyso-VRAC currents were absent. We used this targeting mutant, as well as pharmacological tools, to demonstrate that Lyso-VRAC currents are necessary for the formation of large lysosome-derived vacuoles, which store and then expel excess water to maintain cytosolic water homeostasis. Thus, Lyso-VRACs allow lysosomes of mammalian cells to act as the cell`s "bladder." When Lyso-VRAC current was selectively eliminated, the extent of necrotic cell death to sustained stress was greatly increased, not only in response to hypoosmotic stress, but also to hypoxic and hypothermic stresses. Thus Lyso-VRACs play an essential role in enabling cells to mount successful homeostatic responses to multiple stressors.


Asunto(s)
Lisosomas/metabolismo , Proteínas de la Membrana/metabolismo , Osmorregulación/fisiología , Estrés Fisiológico/fisiología , Animales , Aniones , Células COS , Supervivencia Celular/fisiología , Chlorocebus aethiops , Exocitosis , Técnicas de Inactivación de Genes , Células HEK293 , Homeostasis , Humanos , Proteínas de la Membrana/genética , Ratones , Transcriptoma , Vacuolas
2.
PLoS Biol ; 17(5): e3000252, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31112550

RESUMEN

Rapamycin (Rap) and its derivatives, called rapalogs, are being explored in clinical trials targeting cancer and neurodegeneration. The underlying mechanisms of Rap actions, however, are not well understood. Mechanistic target of rapamycin (mTOR), a lysosome-localized protein kinase that acts as a critical regulator of cellular growth, is believed to mediate most Rap actions. Here, we identified mucolipin 1 (transient receptor potential channel mucolipin 1 [TRPML1], also known as MCOLN1), the principle Ca2+ release channel in the lysosome, as another direct target of Rap. Patch-clamping of isolated lysosomal membranes showed that micromolar concentrations of Rap and some rapalogs activated lysosomal TRPML1 directly and specifically. Pharmacological inhibition or genetic inactivation of mTOR failed to mimic the Rap effect. In vitro binding assays revealed that Rap bound directly to purified TRPML1 proteins with a micromolar affinity. In both healthy and disease human fibroblasts, Rap and rapalogs induced autophagic flux via nuclear translocation of transcription factor EB (TFEB). However, such effects were abolished in TRPML1-deficient cells or by TRPML1 inhibitors. Hence, Rap and rapalogs promote autophagy via a TRPML1-dependent mechanism. Given the demonstrated roles of TRPML1 and TFEB in cellular clearance, we propose that lysosomal TRPML1 may contribute a significant portion to the in vivo neuroprotective and anti-aging effects of Rap via an augmentation of autophagy and lysosomal biogenesis.


Asunto(s)
Lisosomas/metabolismo , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Autofagia/efectos de los fármacos , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Calcio/farmacología , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Células HEK293 , Células HeLa , Humanos , Activación del Canal Iónico/efectos de los fármacos , Lisosomas/efectos de los fármacos , Modelos Biológicos , Unión Proteica/efectos de los fármacos , Sirolimus/análogos & derivados , Sirolimus/química
3.
Phys Chem Chem Phys ; 24(36): 22250-22262, 2022 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-36098073

RESUMEN

Targeting amyloidosis requires high-resolution insight into the underlying mechanisms of amyloid aggregation. The sequence-specific intrinsic properties of a peptide or protein largely govern the amyloidogenic propensity. Thus, it is essential to delineate the structural motifs that define the subsequent downstream amyloidogenic cascade of events. Additionally, it is important to understand the role played by extrinsic factors, such as temperature or sample agitation, in modulating the overall energy barrier that prompts divergent nucleation events. Consequently, these changes can affect the fibrillation kinetics, resulting in structurally and functionally distinct amyloidogenic conformers associated with disease pathogenesis. Here, we have focused on human Islet Polypeptide (hIAPP) amyloidogenesis for the full-length peptide along with its N- and C-terminal fragments, under different temperatures and sample agitation conditions. This helped us to gain a comprehensive understanding of the intrinsic role of specific functional epitopes in the primary structure of the peptide that regulates amyloidogenesis and subsequent cytotoxicity. Intriguingly, our study involving an array of biophysical experiments and ex vivo data suggests a direct influence of external changes on the C-terminal fibrillating sequence. Furthermore, the observations indicate a possible collaborative role of this segment in nucleating hIAPP amyloidogenesis in a physiological scenario, thus making it a potential target for future therapeutic interventions.


Asunto(s)
Amiloidosis , Polipéptido Amiloide de los Islotes Pancreáticos , Amiloide/química , Proteínas Amiloidogénicas , Epítopos , Humanos , Polipéptido Amiloide de los Islotes Pancreáticos/química
4.
Pflugers Arch ; 472(5): 551-560, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32388729

RESUMEN

N-type inactivation of voltage-gated K+ channels is conferred by the N-terminal "ball" domains of select pore-forming α subunits or of auxiliary ß subunits, and influences electrical cellular excitability. Here, we show that hemin impairs inactivation of K+ channels formed by Kv3.4 α subunits as well as that induced by the subunits Kvß1.1, Kvß1.2, and Kvß3.1 when coexpressed with α subunits of the Kv1 subfamily. In Kvß1.1, hemin interacts with cysteine and histidine residues in the N terminus (C7 and H10) with high affinity (EC50 100 nM). Similarly, rapid inactivation of Kv4.2 channels induced by the dipeptidyl peptidase-like protein DPP6a is also sensitive to hemin, and the DPP6a mutation C13S eliminates this dependence. The results suggest a common mechanism for a dynamic regulation of Kv channel inactivation by heme/hemin in N-terminal ball domains of Kv α and auxiliary ß subunits. Free intracellular heme therefore has the potential to regulate cellular excitability via modulation of Kv channel inactivation.


Asunto(s)
Hemina/metabolismo , Activación del Canal Iónico , Canales de Potasio con Entrada de Voltaje/metabolismo , Animales , Sitios de Unión , Dipeptidil-Peptidasas y Tripeptidil-Peptidasas/metabolismo , Células HEK293 , Humanos , Canales de Potasio con Entrada de Voltaje/química , Unión Proteica , Ratas , Xenopus
5.
Int J Mol Sci ; 21(21)2020 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-33167454

RESUMEN

Plants are under constant attack by a suite of insect herbivores. Over millions of years of coexistence, plants have evolved the ability to sense insect feeding via herbivore-associated elicitors in oral secretions, which can mobilize defense responses. However, herbivore-associated elicitors and the intrinsic downstream modulator of such interactions remain less understood. In this study, we show that tobacco hornworm caterpillar (Manduca sexta) oral secretion (OS) induces reactive oxygen species (ROS) in tomato (Solanum lycopersicum) protoplasts. By using a dye-based ROS imaging approach, our study shows that application of plant-fed (PF) M. sexta OS generates significantly higher ROS while artificial diet-fed (DF) caterpillar OS failed to induce ROS in isolated tomato protoplasts. Elevation in ROS generation was saturated after ~140 s of PF OS application. ROS production was also suppressed in the presence of an antioxidant NAC (N-acetyl-L-cysteine). Interestingly, PF OS-induced ROS increase was abolished in the presence of a Ca2+ chelator, BAPTA-AM (1,2-bis(o-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid). These results indicate a potential signaling cascade involving herbivore-associated elicitors, Ca2+, and ROS in plants during insect feeding. In summary, our results demonstrate that plants incorporate a variety of independent signals connected with their herbivores to regulate and mount their defense responses.


Asunto(s)
Manduca/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Solanum lycopersicum/inmunología , Aminoácidos/metabolismo , Animales , Secreciones Corporales/metabolismo , Calcio/metabolismo , Ácido Egtácico/análogos & derivados , Ácido Egtácico/farmacología , Regulación de la Expresión Génica de las Plantas/genética , Herbivoria/fisiología , Larva/metabolismo , Solanum lycopersicum/metabolismo , Manduca/patogenicidad , Protoplastos/inmunología , Protoplastos/metabolismo , Saliva/química , Saliva/metabolismo
6.
Gastroenterology ; 153(6): 1555-1567.e15, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28859856

RESUMEN

BACKGROUND & AIMS: The multiple endocrine neoplasia, type 1 (MEN1) locus encodes the nuclear protein and tumor suppressor menin. MEN1 mutations frequently cause neuroendocrine tumors such as gastrinomas, characterized by their predominant duodenal location and local metastasis at time of diagnosis. Diffuse gastrin cell hyperplasia precedes the appearance of MEN1 gastrinomas, which develop within submucosal Brunner's glands. We investigated how menin regulates expression of the gastrin gene and induces generation of submucosal gastrin-expressing cell hyperplasia. METHODS: Primary enteric glial cultures were generated from the VillinCre:Men1FL/FL:Sst-/- mice or C57BL/6 mice (controls), with or without inhibition of gastric acid by omeprazole. Primary enteric glial cells from C57BL/6 mice were incubated with gastrin and separated into nuclear and cytoplasmic fractions. Cells were incubated with forskolin and H89 to activate or inhibit protein kinase A (a family of enzymes whose activity depends on cellular levels of cyclic AMP). Gastrin was measured in blood, tissue, and cell cultures using an ELISA. Immunoprecipitation with menin or ubiquitin was used to demonstrate post-translational modification of menin. Primary glial cells were incubated with leptomycin b and MG132 to block nuclear export and proteasome activity, respectively. We obtained human duodenal, lymph node, and pancreatic gastrinoma samples, collected from patients who underwent surgery from 1996 through 2007 in the United States or the United Kingdom. RESULTS: Enteric glial cells that stained positive for glial fibrillary acidic protein (GFAP+) expressed gastrin de novo through a mechanism that required PKA. Gastrin-induced nuclear export of menin via cholecystokinin B receptor (CCKBR)-mediated activation of PKA. Once exported from the nucleus, menin was ubiquitinated and degraded by the proteasome. GFAP and other markers of enteric glial cells (eg, p75 and S100B), colocalized with gastrin in human duodenal gastrinomas. CONCLUSIONS: MEN1-associated gastrinomas, which develop in the submucosa, might arise from enteric glial cells through hormone-dependent PKA signaling. This pathway disrupts nuclear menin function, leading to hypergastrinemia and associated sequelae.


Asunto(s)
Duodeno/metabolismo , Gastrinas/metabolismo , Neuroglía/enzimología , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transporte Activo de Núcleo Celular , Animales , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Neoplasias Duodenales/enzimología , Neoplasias Duodenales/genética , Neoplasias Duodenales/patología , Duodeno/efectos de los fármacos , Duodeno/patología , Gastrinoma/enzimología , Gastrinoma/genética , Gastrinoma/patología , Gastrinas/genética , Regulación de la Expresión Génica , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Hiperplasia , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroglía/efectos de los fármacos , Inhibidores de Proteasoma/farmacología , Proteolisis , Proteínas Proto-Oncogénicas/genética , Inhibidores de la Bomba de Protones/farmacología , Receptor de Colecistoquinina B/metabolismo , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Factores de Tiempo , Ubiquitinación
7.
Proc Natl Acad Sci U S A ; 110(42): E4036-44, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24082096

RESUMEN

Fine-tuned regulation of K(+) channel inactivation enables excitable cells to adjust action potential firing. Fast inactivation present in some K(+) channels is mediated by the distal N-terminal structure (ball) occluding the ion permeation pathway. Here we show that Kv1.4 K(+) channels are potently regulated by intracellular free heme; heme binds to the N-terminal inactivation domain and thereby impairs the inactivation process, thus enhancing the K(+) current with an apparent EC50 value of ∼20 nM. Functional studies on channel mutants and structural investigations on recombinant inactivation ball domain peptides encompassing the first 61 residues of Kv1.4 revealed a heme-responsive binding motif involving Cys13:His16 and a secondary histidine at position 35. Heme binding to the N-terminal inactivation domain induces a conformational constraint that prevents it from reaching its receptor site at the vestibule of the channel pore.


Asunto(s)
Hemo , Canal de Potasio Kv1.4 , Animales , Cristalografía por Rayos X , Hemo/química , Hemo/genética , Hemo/metabolismo , Transporte Iónico/fisiología , Canal de Potasio Kv1.4/química , Canal de Potasio Kv1.4/genética , Canal de Potasio Kv1.4/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Xenopus laevis
8.
J Biol Chem ; 287(42): 35565-35575, 2012 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-22927438

RESUMEN

The Kcnh1 gene encodes a voltage-gated potassium channel highly expressed in neurons and involved in tumor cell proliferation, yet its physiological roles remain unclear. We have used the zebrafish as a model to analyze Kcnh1 function in vitro and in vivo. We found that the kcnh1 gene is duplicated in teleost fish (i.e. kcnh1a and kcnh1b) and that both genes are maternally expressed during early development. In adult zebrafish, kcnh1a and kcnh1b have distinct expression patterns but share expression in brain and testis. Heterologous expression of both genes in Xenopus oocytes revealed a strong conservation of characteristic functional properties between human and fish channels, including a unique sensitivity to intracellular Ca(2+)/calmodulin and modulation of voltage-dependent gating by extracellular Mg(2+). Using a morpholino antisense approach, we demonstrate a strong kcnh1 loss-of-function phenotype in developing zebrafish, characterized by growth retardation, delayed hindbrain formation, and embryonic lethality. This late phenotype was preceded by transcriptional up-regulation of known cell-cycle inhibitors (p21, p27, cdh2) and down-regulation of pro-proliferative factors, including cyclin D1, at 70% epiboly. These results reveal an unanticipated basic activity of kcnh1 that is crucial for early embryonic development and patterning.


Asunto(s)
Tipificación del Cuerpo/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Canales de Potasio con Entrada de Voltaje/biosíntesis , Transcripción Genética/fisiología , Proteínas de Pez Cebra/biosíntesis , Pez Cebra/embriología , Animales , Ciclina D1/genética , Ciclina D1/metabolismo , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/genética , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Humanos , Masculino , Especificidad de Órganos/fisiología , Canales de Potasio con Entrada de Voltaje/genética , Rombencéfalo/embriología , Xenopus laevis , Pez Cebra/genética , Proteínas de Pez Cebra/genética
9.
Biochim Biophys Acta ; 1818(5): 1187-95, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22310694

RESUMEN

KCNH1 (EAG1) is a member of the Kv family of voltage-gated potassium channels. However, KCNH1 channels also show some amino-acid sequence similarity to cyclic-nucleotide-regulated channels: they harbor an N-terminal PAS domain, a C-terminal cyclic nucleotide binding homology domain (cNBHD), and N- and C-terminal binding sites for calmodulin. Another notable feature is the channels' high sensitivity toward oxidative modification. Using human KCNH1 expressed in Xenopus oocytes and HEK 293 cells we investigated how oxidative modification alters channel function. Intracellular application of H(2)O(2) or cysteine-specific modifiers potently inhibited KCNH1 channels in two phases. Our systematic cysteine mutagenesis study showed that the rapid and dominant phase was attributed to a right-shift in the voltage dependence of activation, caused by chemical modification of residues C145 and C214. The slow component depended on the C-terminal residues C532 and C562. The cysteine pairs are situated at structural elements linking the transmembrane S1 segment with the PAS domain (N-linker) and the transmembrane channel gate S6 with the cNBH domain (C-linker), respectively. The functional state of KCNH1 channels is determined by the oxidative status of these linkers that provide an additional dimension of channel regulation.


Asunto(s)
Cisteína/metabolismo , Canales de Potasio Éter-A-Go-Go/metabolismo , Activación del Canal Iónico/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Animales , Cisteína/química , Cisteína/genética , Canales de Potasio Éter-A-Go-Go/química , Canales de Potasio Éter-A-Go-Go/genética , Células HEK293 , Humanos , Mutagénesis Sitio-Dirigida , Oxidación-Reducción , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Xenopus laevis
10.
Pathogens ; 11(9)2022 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-36145393

RESUMEN

The purpose of this study was to determine the effect of sanitizer use conditions on the susceptibility, biofilm forming ability and pathogenicity of Listeria monocytogenes. Two different strains of L. monocytogenes and a non-pathogenic L. innocua were exposed to sodium hypochlorite, benzalkonium chloride and peroxyacetic acid at different concentrations (4 to 512 ppm) and treatment times (30 s to 5 min), respectively. Under the tested conditions, no significant difference (p > 0.05) in reduction was observed among the three tested sanitizers. A reduction of 1 to 8 log CFU/mL was observed depending upon the sanitizer concentration and treatment times. The survived cells at the highest sublethal concentration and treatment time of a particular sanitizer upon re-exposure to the same or different sanitizer showed either no change or increased susceptibility when compared to parent strains. Upon repeated exposure to sanitizers at progressively increasing concentrations from 1 to 128 ppm, L. innocua was able to survive concentrations of up to 32 ppm benzalkonium chloride and 64 ppm peroxyacetic acid treatments, respectively. At the tested sub-lethal concentrations, no significant difference (p > 0.05) in biofilm formation was observed among the tested strains. Caco-2 interaction with L. innocua showed a reduction in invasion ability with sublethal concentrations of sanitizers.

11.
Biophys Chem ; 286: 106802, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35605494

RESUMEN

Contact lens wearers are at an increased risk of developing Pseudomonas-associated corneal keratitis, which can lead to a host of serious ocular complications. Despite the use of topical antibiotics, ocular infections remain a major clinical problem, and a strategy to avoid Pseudomonas-associated microbial keratitis is urgently required. The hybrid peptide VR18 (VARGWGRKCPLFGKNKSR) was designed to have enhanced antimicrobial properties in the fight against Pseudomonas-induced microbial keratitis, including contact lens-related keratitis. In this paper, VR18's modes of action against Pseudomonas membranes were shown by live cell Raman spectroscopy, live cell NMR, live-cell fluorescence microscopy and measures taken using sparsely tethered bilayer lipid membrane bacterial models to be via a bacterial-specific membrane disruption mechanism. The high affinity and selectivity of the peptide were then demonstrated using in vivo, in vitro and ex vivo models of Pseudomonas infection. The extensive data presented in this work suggests that topical employment of the VR18 peptide would be a potent therapeutic agent for the prevention or remedy of Pseudomonas-associated microbial keratitis.


Asunto(s)
Antiinfecciosos , Infecciones Bacterianas del Ojo , Queratitis , Antibacterianos/farmacología , Péptidos Antimicrobianos , Infecciones Bacterianas del Ojo/tratamiento farmacológico , Infecciones Bacterianas del Ojo/microbiología , Humanos , Queratitis/tratamiento farmacológico , Queratitis/metabolismo , Queratitis/microbiología , Pseudomonas , Pseudomonas aeruginosa
12.
Sci Rep ; 12(1): 14645, 2022 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-36030326

RESUMEN

Heme, an iron-protoporphyrin IX complex, is a cofactor bound to various hemoproteins and supports a broad range of functions, such as electron transfer, oxygen transport, signal transduction, and drug metabolism. In recent years, there has been a growing recognition of heme as a non-genomic modulator of ion channel functions. Here, we show that intracellular free heme and hemin modulate human ether à go-go (hEAG1, Kv10.1) voltage-gated potassium channels. Application of hemin to the intracellular side potently inhibits Kv10.1 channels with an IC50 of about 4 nM under ambient and 63 nM under reducing conditions in a weakly voltage-dependent manner, favoring inhibition at resting potential. Functional studies on channel mutants and biochemical analysis of synthetic and recombinant channel fragments identified a heme-binding motif CxHx8H in the C-linker region of the Kv10.1 C terminus, with cysteine 541 and histidines 543 and 552 being important for hemin binding. Binding of hemin to the C linker may induce a conformational constraint that interferes with channel gating. Our results demonstrate that heme and hemin are endogenous modulators of Kv10.1 channels and could be exploited to modulate Kv10.1-mediated cellular functions.


Asunto(s)
Canales de Potasio Éter-A-Go-Go , Hemina , Humanos , Potenciales de la Membrana
13.
Chembiochem ; 12(18): 2846-55, 2011 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-22045633

RESUMEN

Studies of the binding of heme/hemin to proteins or peptides have recently intensified as it became evident that heme serves not only as a prosthetic group, but also as a regulator and effector molecule interacting with transmembrane and cytoplasmic proteins. The iron-ion-containing heme group can associate with these proteins in different ways, with the amino acids Cys, His, and Tyr allowing individual modes of binding. Strong coordinate-covalent binding, such as in cytochrome c, is known, and reversible attachment is also discussed. Ligands for both types of binding have been reported independently, though sometimes with different affinities for similar sequences. We applied a combinatorial approach using the library (X)(4) (C/H/Y)(X)(4) to characterize peptide ligands with considerable hemin binding capacities. Some of the library-selected peptides were comparable in terms of hemin association independently of whether or not a cysteine residue was present in the sequence. Indeed, a preference for His-based (≈39 %) and Tyr-based (≈40 %) sequences over Cys-based ones (≈21 %) was detected. The binding affinities for the library-selected peptides, as determined by UV/Vis spectroscopy, were in the nanomolar range. Moreover, selected representatives efficiently competed for hemin binding with the human BK channel hSlo1, which is known to be regulated by heme through binding to its heme-binding domain.


Asunto(s)
Técnicas Químicas Combinatorias , Hemina/metabolismo , Biblioteca de Péptidos , Proteínas/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Unión Proteica , Proteínas/química , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Espectrofotometría Ultravioleta
14.
Cells ; 10(9)2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34571868

RESUMEN

Plants and insect herbivores are in a relentless battle to outwit each other. Plants have evolved various strategies to detect herbivores and mount an effective defense system against them. These defenses include physical and structural barriers such as spines, trichomes, cuticle, or chemical compounds, including secondary metabolites such as phenolics and terpenes. Plants perceive herbivory by both mechanical and chemical means. Mechanical sensing can occur through the perception of insect biting, piercing, or chewing, while chemical signaling occurs through the perception of various herbivore-derived compounds such as oral secretions (OS) or regurgitant, insect excreta (frass), or oviposition fluids. Interestingly, ion channels or transporters are the first responders for the perception of these mechanical and chemical cues. These transmembrane pore proteins can play an important role in plant defense through the induction of early signaling components such as plasma transmembrane potential (Vm) fluctuation, intracellular calcium (Ca2+), and reactive oxygen species (ROS) generation, followed by defense gene expression, and, ultimately, plant defense responses. In recent years, studies on early plant defense signaling in response to herbivory have been gaining momentum with the application of genetically encoded GFP-based sensors for real-time monitoring of early signaling events and genetic tools to manipulate ion channels involved in plant-herbivore interactions. In this review, we provide an update on recent developments and advances on early signaling events in plant-herbivore interactions, with an emphasis on the role of ion channels in early plant defense signaling.


Asunto(s)
Herbivoria/fisiología , Insectos/metabolismo , Canales Iónicos/metabolismo , Transducción de Señal/fisiología , Animales , Humanos , Potenciales de la Membrana/fisiología , Plantas/metabolismo
15.
Front Plant Sci ; 12: 745891, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35295863

RESUMEN

Sugarcane (Saccharum spp.) is a prominent source of sugar and serves as bioenergy/biomass feedstock globally. Multiple biotic and abiotic stresses, including drought, salinity, and cold, adversely affect sugarcane yield. G-protein-coupled receptors (GPCRs) are components of G-protein-mediated signaling affecting plant growth, development, and stress responses. Here, we identified a GPCR-like protein (ShGPCR1) from sugarcane and energy cane (Saccharum spp. hybrids) and characterized its function in conferring tolerance to multiple abiotic stresses. ShGPCR1 protein sequence contained nine predicted transmembrane (TM) domains connected by four extracellular and four intracellular loops, which could interact with various ligands and heterotrimeric G proteins in the cells. ShGPCR1 sequence displayed other signature features of a GPCR, such as a putative guanidine triphosphate (GTP)-binding domain, as well as multiple myristoylation and protein phosphorylation sites, presumably important for its biochemical function. Expression of ShGPCR1 was upregulated by drought, salinity, and cold stresses. Subcellular imaging and calcium (Ca2+) measurements revealed that ShGPCR1 predominantly localized to the plasma membrane and enhanced intracellular Ca2+ levels in response to GTP, respectively. Furthermore, constitutive overexpression of ShGPCR1 in sugarcane conferred tolerance to the three stressors. The stress-tolerance phenotype of the transgenic lines corresponded with activation of multiple drought-, salinity-, and cold-stress marker genes, such as Saccharum spp. LATE EMBRYOGENESIS ABUNDANT, DEHYDRIN, DROUGHT RESPONSIVE 4, GALACTINOL SYNTHASE, ETHYLENE RESPONSIVE FACTOR 3, SALT OVERLY SENSITIVE 1, VACUOLAR Na+/H+ ANTIPORTER 1, NAM/ATAF1/2/CUC2, COLD RESPONSIVE FACTOR 2, and ALCOHOL DEHYDROGENASE 3. We suggest that ShGPCR1 plays a key role in conferring tolerance to multiple abiotic stresses, and the engineered lines may be useful to enhance sugarcane production in marginal environments with fewer resources.

16.
J Physiol ; 588(Pt 16): 2999-3009, 2010 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-20547678

RESUMEN

Excess reactive oxygen species (ROS) play a crucial role under pathophysiological conditions, such as ischaemia/reperfusion and diabetes, potentially contributing to cardiac arrhythmia. hERG1 (KCNH2) potassium channels terminate the cardiac action potential and malfunction can lead to long-QT syndrome and fatal arrhythmia. To investigate the molecular mechanisms of hERG1 channel alteration by ROS, hERG1 and mutants thereof were expressed in HEK293 cells and studied with the whole-cell patch-clamp method. Even mild ROS stress induced by hyperglycaemia markedly decreased channel current. Intracellular H2O2 or cysteine-specific modifiers also strongly inhibited channel activity and accelerated deactivation kinetics. Mutagenesis revealed that cysteine 723 (C723), a conserved residue in a structural element linking the C-terminal domain to the channel's gate, is critical for oxidative functional modification. Moreover, kinetics of channel closure strongly influences ROS-induced modification, where rapid channel deactivation diminishes ROS sensitivity. Because of its fast deactivation kinetics, the N-terminally truncated splice variant hERG1b possesses greater resistance to oxidative modification.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/metabolismo , Activación del Canal Iónico , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Línea Celular , Cisteína , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/química , Canales de Potasio Éter-A-Go-Go/genética , Glucosa/metabolismo , Humanos , Cinética , Potenciales de la Membrana , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Oxidantes/farmacología , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad , Reactivos de Sulfhidrilo/farmacología , Transfección
17.
Methods Mol Biol ; 2115: 365-383, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32006411

RESUMEN

CRISPR Cas9 genome editing allows researchers to modify genes in a multitude of ways including to obtain deletions, epitope-tagged loci, and knock-in mutations. Within 6 years of its initial application, CRISPR-Cas9 genome editing has been widely employed, but disadvantages to this method, such as low modification efficiencies and off-target effects, need careful consideration. Obtaining custom donor vectors can also be expensive and time-consuming. This chapter details strategies to overcome barriers to CRISPR-Cas9 genome editing as well as recent developments in employing this technique.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica/métodos , Proteína 9 Asociada a CRISPR/genética , Línea Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Proteína Forkhead Box O3/genética , Vectores Genéticos/genética , Humanos , Mutación , ARN Guía de Kinetoplastida/genética
18.
ACS Chem Neurosci ; 11(13): 1965-1977, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32492332

RESUMEN

Alzheimer's disease (AD) is a severe neurodegenerative disorder caused by abnormal accumulation of toxic amyloid plaques of the amyloid-beta (Aß) or the tau proteins in the brain. The plaque deposition leading to the collapse of the cellular integrity is responsible for a myriad of surface phenomena acting at the neuronal lipid interface. Recent years have witnessed dysfunction of the blood-brain barriers (BBB) associated with AD. Several studies support the idea that BBB acts as a platform for the formation of misfolded Aß peptide, promoting oligomerization and fibrillation, compromising the overall integrity of the central nervous system. While the amyloid plaque deposition has been known to be responsible for the collapse of the BBB membrane integrity, the causal effect relationship between BBB and Aß amyloidogenesis remains unclear. In this study, we have used physiologically relevant synthetic model membrane systems to gain atomic insight into the functional aspects of the lipid interface. Here, we have used a minimalist BBB mimic, POPC/POPG/cholesterol/GM1, to compare with the native BBB (total lipid brain extract (TLBE)), to understand the molecular events occurring in the membrane-induced Aß40 amyloid aggregation. Our study showed that the two membrane models accelerated the Aß40 aggregation kinetics with differential secondary structural transitions of the peptide. The observed structural transitions are defined by the lipid compositions, which in turn undermines the differences in lipid surface phenomena, leading to peptide induced cellular toxicity in the neuronal membrane.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Amiloide/metabolismo , Péptidos beta-Amiloides/metabolismo , Barrera Hematoencefálica/metabolismo , Humanos , Placa Amiloide
19.
Sci Adv ; 6(6): eaaz2736, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32128386

RESUMEN

Duchenne muscular dystrophy (DMD) is a devastating disease caused by mutations in dystrophin that compromise sarcolemma integrity. Currently, there is no treatment for DMD. Mutations in transient receptor potential mucolipin 1 (ML1), a lysosomal Ca2+ channel required for lysosomal exocytosis, produce a DMD-like phenotype. Here, we show that transgenic overexpression or pharmacological activation of ML1 in vivo facilitates sarcolemma repair and alleviates the dystrophic phenotypes in both skeletal and cardiac muscles of mdx mice (a mouse model of DMD). Hallmark dystrophic features of DMD, including myofiber necrosis, central nucleation, fibrosis, elevated serum creatine kinase levels, reduced muscle force, impaired motor ability, and dilated cardiomyopathies, were all ameliorated by increasing ML1 activity. ML1-dependent activation of transcription factor EB (TFEB) corrects lysosomal insufficiency to diminish muscle damage. Hence, targeting lysosomal Ca2+ channels may represent a promising approach to treat DMD and related muscle diseases.


Asunto(s)
Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Canales de Potencial de Receptor Transitorio/agonistas , Animales , Biomarcadores , Biopsia , Modelos Animales de Enfermedad , Distrofina/genética , Técnica del Anticuerpo Fluorescente , Expresión Génica , Ratones , Ratones Endogámicos mdx , Ratones Transgénicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Distrofia Muscular de Duchenne/tratamiento farmacológico , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/patología , Miocardio/metabolismo , Miocardio/patología
20.
FEBS J ; 275(19): 4756-66, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18721141

RESUMEN

The type II fatty acid synthase (FAS) pathway of Plasmodium falciparum is a validated unique target for developing novel antimalarials, due to its intrinsic differences from the typeI pathway operating in humans. beta-Ketoacyl acyl carrier protein (ACP) reductase (FabG) performs the NADPH-dependent reduction of beta-ketoacyl-ACP to beta-hydroxyacyl-ACP, the first reductive step in the elongation cycle of fatty acid biosynthesis. In this article, we report intensive studies on the direct interactions of Plasmodium FabG and Plasmodium ACP in solution, in the presence and absence of its cofactor, NADPH, by monitoring the change in intrinsic fluorescence of P.falciparum FabG (PfFabG) and by surface plasmon resonance. To address the issue of the importance of the residues involved in strong, specific and stoichiometric binding of PfFabG to P.falciparum ACP (PfACP), we mutated Arg187, Arg190 and Arg230 of PfFabG. The activities of the mutants were assessed using both an ACP-dependent and an ACP-independent assay. The affinities of all the PfFabG mutants for acetoacetyl-ACP (the physiological substrate) were reduced to different extents as compared to wild-type PfFabG, but were equally active in biochemical assays with the substrate analog acetoacetyl-CoA. Kinetic analysis and studies of direct binding between PfFabG and PfACP confirmed the identification of Arg187 and Arg230 as critical residues for the PfFabG-PfACP interactions. Our studies thus reveal the significance of the positively charged/hydrophobic patch located adjacent to the active site cavities of PfFabG for interactions with PfACP.


Asunto(s)
Proteína Transportadora de Acilo/metabolismo , Oxidorreductasas de Alcohol/química , Oxidorreductasas de Alcohol/genética , Plasmodium falciparum/enzimología , 3-Oxoacil-(Proteína Transportadora de Acil) Reductasa , Oxidorreductasas de Alcohol/metabolismo , Secuencia de Aminoácidos , Animales , Arginina/metabolismo , Sitios de Unión , Cinética , Datos de Secuencia Molecular , NADP/metabolismo , Plasmodium falciparum/metabolismo , Alineación de Secuencia , Resonancia por Plasmón de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA