Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell ; 187(9): 2224-2235.e16, 2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38614101

RESUMEN

The membrane protein NINJ1 mediates plasma membrane rupture in pyroptosis and other lytic cell death pathways. Here, we report the cryo-EM structure of a NINJ1 oligomer segmented from NINJ1 rings. Each NINJ1 subunit comprises amphipathic (⍺1, ⍺2) and transmembrane (TM) helices (⍺3, ⍺4) and forms a chain of subunits, mainly by the TM helices and ⍺1. ⍺3 and ⍺4 are kinked, and the Gly residues are important for function. The NINJ1 oligomer possesses a concave hydrophobic side that should face the membrane and a convex hydrophilic side formed by ⍺1 and ⍺2, presumably upon activation. This structural observation suggests that NINJ1 can form membrane disks, consistent with membrane fragmentation by recombinant NINJ1. Live-cell and super-resolution imaging uncover ring-like structures on the plasma membrane that are released into the culture supernatant. Released NINJ1 encircles a membrane inside, as shown by lipid staining. Therefore, NINJ1-mediated membrane disk formation is different from gasdermin-mediated pore formation, resulting in membrane loss and plasma membrane rupture.


Asunto(s)
Moléculas de Adhesión Celular Neuronal , Membrana Celular , Microscopía por Crioelectrón , Membrana Celular/metabolismo , Humanos , Moléculas de Adhesión Celular Neuronal/metabolismo , Moléculas de Adhesión Celular Neuronal/química , Animales , Ratones , Células HEK293 , Piroptosis , Modelos Moleculares , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/química , Proteínas de Unión a Fosfato/metabolismo
2.
Transl Res ; 267: 39-53, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38042478

RESUMEN

General anesthetic drugs cause cognitive deficits that persist after the drugs have been eliminated. Astrocytes may contribute to such cognition-impairing effects through the release of one or more paracrine factors that increase a tonic inhibitory conductance generated by extrasynaptic γ-aminobutyric acid type A (GABAA) receptors in hippocampal neurons. The mechanisms underlying this astrocyte-to-neuron crosstalk remain unknown. Interestingly, astrocytes express anesthetic-sensitive GABAA receptors. Here, we tested the hypothesis that anesthetic drugs activate astrocytic GABAA receptors to initiate crosstalk leading to a persistent increase in extrasynaptic GABAA receptor function in neurons. We also investigated the signaling pathways in neurons and aimed to identify the paracrine factors released from astrocytes. Astrocytes and neurons from mice were grown in primary cell cultures and studied using in vitro electrophysiological and biochemical assays. We discovered that the commonly used anesthetics etomidate (injectable) and sevoflurane (inhaled) stimulated astrocytic GABAA receptors, which in turn promoted the release paracrine factors, that increased the tonic current in neurons via a p38 MAPK-dependent signaling pathway. The increase in tonic current was mimicked by exogenous IL-1ß and abolished by blocking IL-1 receptors; however, unexpectedly, IL-1ß and other cytokines were not detected in astrocyte-conditioned media. In summary, we have identified a novel form of crosstalk between GABAA receptors in astrocytes and neurons that engages a p38 MAPK-dependent pathway. Brief commentary BACKGROUND: Many older patients experience cognitive deficits after surgery. Anesthetic drugs may be a contributing factor as they cause a sustained increase in the function of "memory blocking" extrasynaptic GABAA receptors in neurons. Interestingly, astrocytes are required for this increase; however, the mechanisms underlying the astrocyte-to-neuron crosstalk remain unknown. TRANSLATIONAL SIGNIFICANCE: We discovered that commonly used general anesthetic drugs stimulate GABAA receptors in astrocytes, which in turn release paracrine factors that trigger a persistent increase in extrasynaptic GABAA receptor function in neurons via p38 MAPK. This novel form of crosstalk may contribute to persistent cognitive deficits after general anesthesia and surgery.


Asunto(s)
Anestésicos Generales , Receptores de GABA-A , Humanos , Ratones , Animales , Receptores de GABA-A/metabolismo , Astrocitos/metabolismo , Neuronas , Anestésicos Generales/farmacología , Ácido gamma-Aminobutírico/metabolismo , Ácido gamma-Aminobutírico/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
3.
Curr Biol ; 33(7): 1282-1294.e5, 2023 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-36898371

RESUMEN

The ongoing metabolic and microbicidal pathways that support and protect cellular life generate potentially damaging reactive oxygen species (ROS). To counteract damage, cells express peroxidases, which are antioxidant enzymes that catalyze the reduction of oxidized biomolecules. Glutathione peroxidase 4 (GPX4) is the major hydroperoxidase specifically responsible for reducing lipid peroxides; this homeostatic mechanism is essential, and its inhibition causes a unique type of lytic cell death, ferroptosis. The mechanism(s) that lead to cell lysis in ferroptosis, however, are unclear. We report that the lipid peroxides formed during ferroptosis accumulate preferentially at the plasma membrane. Oxidation of surface membrane lipids increased tension on the plasma membrane and led to the activation of Piezo1 and TRP channels. Oxidized membranes thus became permeable to cations, ultimately leading to the gain of cellular Na+ and Ca2+ concomitant with loss of K+. These effects were reduced by deletion of Piezo1 and completely inhibited by blocking cation channel conductance with ruthenium red or 2-aminoethoxydiphenyl borate (2-APB). We also found that the oxidation of lipids depressed the activity of the Na+/K+-ATPase, exacerbating the dissipation of monovalent cation gradients. Preventing the changes in cation content attenuated ferroptosis. Altogether, our study establishes that increased membrane permeability to cations is a critical step in the execution of ferroptosis and identifies Piezo1, TRP channels, and the Na+/K+-ATPase as targets/effectors of this type of cell death.


Asunto(s)
Ferroptosis , Peróxidos Lipídicos , Cationes , Glutatión Peroxidasa/metabolismo , Peroxidación de Lípido/fisiología , Peróxidos Lipídicos/metabolismo , Fosfolípido Hidroperóxido Glutatión Peroxidasa/metabolismo , Proteínas de la Membrana/metabolismo
4.
Elife ; 112022 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-36468682

RESUMEN

First recognized more than 30 years ago, glycine protects cells against rupture from diverse types of injury. This robust and widely observed effect has been speculated to target a late downstream process common to multiple modes of tissue injury. The molecular target of glycine that mediates cytoprotection, however, remains elusive. Here, we show that glycine works at the level of NINJ1, a newly identified executioner of plasma membrane rupture in pyroptosis, necrosis, and post-apoptosis lysis. NINJ1 is thought to cluster within the plasma membrane to cause cell rupture. We demonstrate that the execution of pyroptotic cell rupture is similar for human and mouse NINJ1 and that NINJ1 knockout functionally and morphologically phenocopies glycine cytoprotection in macrophages undergoing lytic cell death. Next, we show that glycine prevents NINJ1 clustering by either direct or indirect mechanisms. In pyroptosis, glycine preserves cellular integrity but does not affect upstream inflammasome activities or accompanying energetic cell death. By positioning NINJ1 clustering as a glycine target, our data resolve a long-standing mechanism for glycine-mediated cytoprotection. This new understanding will inform the development of cell preservation strategies to counter pathologic lytic cell death.


Asunto(s)
Glicina , Piroptosis , Ratones , Humanos , Animales , Glicina/farmacología , Glicina/metabolismo , Muerte Celular , Inflamasomas/metabolismo , Membrana Celular/metabolismo , Análisis por Conglomerados , Moléculas de Adhesión Celular Neuronal/metabolismo , Factores de Crecimiento Nervioso/metabolismo
5.
Endocr Connect ; 10(8): 861-872, 2021 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-34319253

RESUMEN

Plasma free fatty acids (FFAs) are elevated in obesity and can induce insulin resistance via endoplasmic reticulum (ER) stress. However, it is unknown whether hepatic insulin resistance caused by the elevation of plasma FFAs is alleviated by chemical chaperones. Rats received one of the following i.v. treatments for 48 h: saline, intralipid plus heparin (IH), IH plus the chemical chaperone 4-phenylbutyric acid (PBA), or PBA alone and a hyperinsulinemic-euglycemic clamp was performed during the last 2 h. PBA co-infusion normalized IH-induced peripheral insulin resistance, similar to our previous findings with an antioxidant and an IκBα kinase ß (IKKß) inhibitor. Different from our previous results with the antioxidant and IKKß inhibitor, PBA also improved IH-induced hepatic insulin resistance in parallel with activation of Akt. Unexpectedly, IH did not induce markers of ER stress in the liver, but PBA prevented IH-induced elevation of phosphorylated eukaryotic initiation factor-2α protein in adipose tissue. PBA tended to decrease circulating fetuin-A and significantly increased circulating fibroblast growth factor 21 (FGF21) without affecting markers of activation of hepatic protein kinase C-δ or p38 mitogen-activated protein kinase that we have previously involved in hepatic insulin resistance in this model. In conclusion: (i) PBA prevented hepatic insulin resistance caused by prolonged plasma FFA elevation without affecting hepatic ER stress markers; (ii) the PBA effect is likely due to increased FGF21 and/or decreased fetuin-A, which directly signal to upregulate Akt activation.

6.
J Cell Sci ; 134(7)2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33722976

RESUMEN

Rab5 is required for macropinosome formation, but its site and mode of action remain unknown. We report that Rab5 acts at the plasma membrane, downstream of ruffling, to promote macropinosome sealing and scission. Dominant-negative Rab5, which obliterates macropinocytosis, had no effect on the development of membrane ruffles. However, Rab5-containing vesicles were recruited to circular membrane ruffles, and soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE)-dependent endomembrane fusion was necessary for the completion of macropinocytosis. This fusion event coincided with the disappearance of PtdIns(4,5)P2 that accompanies macropinosome closure. Counteracting the depletion of PtdIns(4,5)P2 by expression of phosphatidylinositol-4-phosphate 5-kinase impaired macropinosome formation. Importantly, we found that the removal of PtdIns(4,5)P2 is dependent on Rab5, through the Rab5-mediated recruitment of the inositol 5-phosphatases OCRL and Inpp5b, via APPL1. Knockdown of OCRL and Inpp5b, or APPL1, prevented macropinosome closure without affecting ruffling. We therefore propose that Rab5 is essential for the clearance of PtdIns(4,5)P2 needed to complete the scission of macropinosomes or to prevent their back-fusion with the plasmalemma.


Asunto(s)
Fosfatidilinositol 4,5-Difosfato , Fosfatidilinositoles , Inositol , Inositol Polifosfato 5-Fosfatasas , Pinocitosis
7.
Nat Commun ; 11(1): 4561, 2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32917873

RESUMEN

The protein high-mobility group box 1 (HMGB1) is released into the extracellular space in response to many inflammatory stimuli, where it is a potent signaling molecule. Although research has focused on downstream HMGB1 signaling, the means by which HMGB1 exits the cell is controversial. Here we demonstrate that HMGB1 is not released from bone marrow-derived macrophages (BMDM) after lipopolysaccharide (LPS) treatment. We also explore whether HMGB1 is released via the pore-forming protein gasdermin D after inflammasome activation, as is the case for IL-1ß. HMGB1 is only released under conditions that cause cell lysis (pyroptosis). When pyroptosis is prevented, HMGB1 is not released, despite inflammasome activation and IL-1ß secretion. During endotoxemia, gasdermin D knockout mice secrete HMGB1 normally, yet secretion of IL-1ß is completely blocked. Together, these data demonstrate that in vitro HMGB1 release after inflammasome activation occurs after cellular rupture, which is probably inflammasome-independent in vivo.


Asunto(s)
Proteína HMGB1/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Animales , Modelos Animales de Enfermedad , Endotoxemia/metabolismo , Femenino , Proteína HMGB1/genética , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Lipopolisacáridos/efectos adversos , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Unión a Fosfato/genética , Piroptosis , Transducción de Señal
8.
Sci Adv ; 6(14): eaay9572, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32270040

RESUMEN

The endoplasmic reticulum (ER) is a highly dynamic network of membranes. Here, we combine live-cell microscopy with in situ cryo-electron tomography to directly visualize ER dynamics in several secretory cell types including pancreatic ß-cells and neurons under near-native conditions. Using these imaging approaches, we identify a novel, mobile form of ER, ribosome-associated vesicles (RAVs), found primarily in the cell periphery, which is conserved across different cell types and species. We show that RAVs exist as distinct, highly dynamic structures separate from the intact ER reticular architecture that interact with mitochondria via direct intermembrane contacts. These findings describe a new ER subcompartment within cells.


Asunto(s)
Vesículas Citoplasmáticas/metabolismo , Retículo Endoplásmico/metabolismo , Ribosomas/metabolismo , Animales , Transporte Biológico , Microscopía por Crioelectrón , Vesículas Citoplasmáticas/ultraestructura , Retículo Endoplásmico/ultraestructura , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Ratones , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Imagen Molecular , Especificidad de Órganos , Ratas , Ribosomas/ultraestructura , Estrés Fisiológico
10.
Endocrinology ; 159(11): 3643-3654, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30215691

RESUMEN

Prolonged elevation of glucose can adversely affect ß-cell function. Oxidative stress, which has been implicated in glucose-induced ß-cell dysfunction, can activate c-jun N-terminal kinase (JNK). However, whether JNK is causal in glucose-induced ß-cell dysfunction in vivo is unclear. Therefore, we aimed at investigating the causal role of JNK activation in in vivo models of glucose-induced ß-cell dysfunction. Glucose-induced ß-cell dysfunction was investigated in the presence or absence of JNK inhibition. JNK inhibition was achieved using either (i) the JNK-specific inhibitor SP600125 or (ii) JNK-1-null mice. (i) Rats or mice were infused intravenously with saline or glucose with or without SP600125. (ii) JNK-1 null mice and their littermate wild-type controls were infused intravenously with saline or glucose. Following the glucose infusion periods in rats and mice, ß-cell function was assessed in isolated islets or in vivo using hyperglycemic clamps. Forty-eight-hour hyperglycemia at ~20 mM in rats or 96-hour hyperglycemia at ~13 mM in mice impaired ß-cell function in isolated islets and in vivo. Inhibition of JNK using either SP600125 or JNK-1-null mice prevented glucose-induced ß-cell dysfunction in isolated islets and in vivo. Islets of JNK-1-null mice exposed to hyperglycemia in vivo showed an increase in Pdx-1 and insulin 2 mRNA, whereas islets of wild-type mice did not. Together, these data show that JNK pathway is involved in glucose-induced ß-cell dysfunction in vivo and is thus a potential therapeutic target for type 2 diabetes.


Asunto(s)
Antracenos/farmacología , Glucosa/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 8 Activada por Mitógenos/genética , Inhibidores de Proteínas Quinasas/farmacología , Animales , Diabetes Mellitus Tipo 2/metabolismo , Técnica de Clampeo de la Glucosa , Proteínas de Homeodominio/efectos de los fármacos , Proteínas de Homeodominio/genética , Hiperglucemia/metabolismo , Técnicas In Vitro , Insulina/genética , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Noqueados , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Transducción de Señal , Transactivadores/efectos de los fármacos , Transactivadores/genética
11.
Eur Respir J ; 51(4)2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29519920

RESUMEN

Human mesenchymal stem/stromal cells (MSCs) have been reported to produce an M2-like, alternatively activated phenotype in macrophages. In addition, MSCs mediate effective bacterial clearance in pre-clinical sepsis models. Thus, MSCs have a paradoxical antimicrobial and anti-inflammatory response that is not understood.Here, we studied the phenotypic and functional response of monocyte-derived human macrophages to MSC exposure in vitroMSCs induced two distinct, coexistent phenotypes: M2-like macrophages (generally elongated morphology, CD163+, acute phagosomal acidification, low NOX2 expression and limited phagosomal superoxide production) and M1-like macrophages characterised by high levels of phagosomal superoxide production. Enhanced phagosomal reactive oxygen species production was also observed in alveolar macrophages from a rodent model of pneumonia-induced sepsis. The production of M1-like macrophages was dependent on prostaglandin E2 and phosphatidylinositol 3-kinase. MSCs enhanced human macrophage phagocytosis of unopsonised bacteria and enhanced bacterial killing compared with untreated macrophages. Bacterial killing was significantly reduced by blockade of NOX2 using diphenyleneiodonium, suggesting that M1-like cells are primarily responsible for this effect. MSCs also enhanced phagocytosis and polarisation of M1-like macrophages derived from patients with severe sepsis.The enhanced antimicrobial capacity (M1-like) and inflammation resolving phenotype (M2-like) may account for the paradoxical effect of these cells in sepsis in vivo.


Asunto(s)
Infecciones por Escherichia coli/inmunología , Macrófagos Alveolares/citología , Células Madre Mesenquimatosas/citología , NADPH Oxidasa 2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sepsis/inmunología , Animales , Diferenciación Celular , Técnicas de Cocultivo , Humanos , Activación de Macrófagos , Macrófagos Alveolares/microbiología , Células Madre Mesenquimatosas/microbiología , Fagocitosis , Ratas Sprague-Dawley
12.
J Endocr Soc ; 2(4): 336-347, 2018 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-29577109

RESUMEN

Nod-like receptor (NLR)X1 is an NLR family protein that localizes to the mitochondrial matrix and modulates reactive oxygen species production, possibly by directly interacting with the electron transport chain. Recent work demonstrated that cells lacking NLRX1 have higher oxygen consumption but lower levels of adenosine triphosphate, suggesting that NLRX1 might prevent uncoupling of oxidative phosphorylation. We therefore hypothesized that NLRX1 might regulate whole-body energy metabolism through its effect on mitochondria. Male NLRX1 whole-body knockout (KO) mice and wild-type (WT) C57BL/6N controls were fed a low-fat or a high-fat (HF) diet for 16 weeks from weaning. Contrary to this hypothesis, there were no differences in body weight, adiposity, energy intake, or energy expenditure between HF-fed KO and WT mice, but instead HF KO mice were partially protected from the development of diet-induced hyperglycemia. Additionally, HF KO mice did not present with hyperinsulinemia during the glucose tolerance test, as did HF WT mice. There were no genotype differences in insulin tolerance, which led us to consider a pancreatic phenotype. Histology revealed that KO mice were protected from HF-induced pancreatic lipid accumulation, suggesting a potential role for NLRX1 in pancreatic dysfunction during the development diet-induced type 2 diabetes mellitus. Hence, NLRX1 depletion partially protects against postabsorptive hyperglycemia in obesity that may be linked to the prevention of pancreatic lipid accumulation. Although the actual mechanisms restoring glucose and insulin dynamics remain unknown, NLRX1 emerges as a potentially interesting target to inhibit for the prevention of type 2 diabetes mellitus.

13.
Diabetologia ; 60(10): 2021-2032, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28725915

RESUMEN

AIMS/HYPOTHESIS: We have previously shown that oxidative stress plays a causal role in beta cell dysfunction induced by fat. Here, we address whether the proinflammatory kinase inhibitor of (nuclear factor) κB kinase ß (IKKß), which is activated by oxidative stress, is also implicated. METHODS: Fat (oleate or olive oil) was infused intravenously in Wistar rats for 48 h with or without the IKKß inhibitor salicylate. Thereafter, beta cell function was evaluated in vivo using hyperglycaemic clamps or ex vivo in islets isolated from fat-treated rats. We also exposed rat islets to oleate in culture, with or without salicylate and 4(2'-aminoethyl)amino-1,8-dimethylimidazo(1,2-a)quinoxaline; BMS-345541 (BMS, another inhibitor of IKKß) and evaluated beta cell function in vitro. Furthermore, oleate was infused in mice treated with BMS and in beta cell-specific Ikkb-null mice. RESULTS: 48 h infusion of fat impaired beta-cell function in vivo, assessed using the disposition index (DI), in rats (saline: 1.41 ± 0.13; oleate: 0.95 ± 0.11; olive oil [OLO]: 0.87 ± 0.15; p < 0.01 for both fats vs saline) and in mice (saline: 2.51 ± 0.39; oleate: 1.20 ± 0.19; p < 0.01 vs saline) and ex vivo (i.e., insulin secretion, units are pmol insulin islet-1 h-1) in rat islets (saline: 1.51 ± 0.13; oleate: 1.03 ± 0.10; OLO: 0.91 ± 0.13; p < 0.001 for both fats vs saline) and the dysfunction was prevented by co-infusion of salicylate in rats (oleate + salicylate: 1.30 ± 0.09; OLO + salicylate: 1.33 ± 0.23) or BMS in mice (oleate + BMS: 2.25 ± 0.42) in vivo and by salicylate in rat islets ex vivo (oleate + salicylate: 1.74 ± 0.31; OLO + salicylate: 1.54 ± 0.29). In cultured islets, 48 h exposure to oleate impaired beta-cell function ([in pmol insulin islet-1 h-1] control: 0.66 ± 0.12; oleate: 0.23 ± 0.03; p < 0.01 vs saline), an effect prevented by both inhibitors (oleate + salicylate: 0.98 ± 0.08; oleate + BMS: 0.50 ± 0.02). Genetic inhibition of IKKß also prevented fat-induced beta-cell dysfunction ex vivo ([in pmol insulin islet-1 h-1] control saline: 0.16 ± 0.02; control oleate: 0.10 ± 0.02; knockout oleate: 0.17 ± 0.04; p < 0.05 control saline vs. control oleate) and in vivo (DI: control saline: 3.86 ± 0.40; control oleate: 1.95 ± 0.29; knockout oleate: 2.96 ± 0.24; p < 0.01 control saline vs control oleate). CONCLUSIONS/INTERPRETATION: Our results demonstrate a causal role for IKKß in fat-induced beta cell dysfunction in vitro, ex vivo and in vivo.


Asunto(s)
Ácidos Grasos no Esterificados/metabolismo , Quinasa I-kappa B/antagonistas & inhibidores , Células Secretoras de Insulina/efectos de los fármacos , Ácido Oléico/farmacología , Ácido Salicílico/farmacología , Animales , Femenino , Imidazoles/farmacología , Células Secretoras de Insulina/metabolismo , Ratones , Estrés Oxidativo/efectos de los fármacos , Quinoxalinas/farmacología , Ratas , Ratas Wistar
14.
Am J Med Genet A ; 173(7): 1782-1791, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28475290

RESUMEN

Recurrent microduplications/microdeletions of 1q21.1 are characterized by variable phenotypes ranging from normal development to developmental delay (DD) and congenital anomalies. Their interpretation is challenging especially in families with affected and unaffected carriers. We used whole exome sequencing (WES) to look for sequence variants in two male probands with inherited 1q21.1 CNVs that could explain their more severe phenotypes. One proband had a 1q21.1 deletion transmitted from maternal grandmother, while the other had a paternal duplication. We found mutations in five genes (SMPD1, WNK3, NOS1, ATF6, and EFHC1) that could contribute to the more severe phenotype in the probands in comparison to their mildly affected or unaffected 1q21.1 CNV carrying relatives. Interestingly, all genes have roles in stress responses (oxidative/Endoplasmic Reticulum (ER)/osmotic). One of the variants was in an X-linked gene WNK3 and segregated with the developmental features and X inactivation pattern in the family with 1q21.1 deletion transmitted from maternal grandmother. In silico analysis of all rare deleterious variants in both probands identified enrichment in nervous system diseases, metabolic pathways, protein processing in the ER and protein export. Our studies suggest that rare deleterious variants outside of the 1q21.1 CNV, individually or as a pool, could contribute to phenotypic variability in carriers of this CNV. Rare deleterious variants in stress response genes are of interest and raise the possibility of susceptibility of carriers to variable environmental influences. Next generation sequencing of additional familial cases with 1q21.1 CNV could further help determine the possible causes of phenotypic variability in carriers of this CNV.

15.
Lab Chip ; 16(15): 2921-34, 2016 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-27378588

RESUMEN

Tissues are challenging to genetically manipulate due to limited penetration of viral particles resulting in low transduction efficiency. We are particularly interested in expressing genetically-encoded sensors in ex vivo pancreatic islets to measure glucose-stimulated metabolism, however poor viral penetration biases these measurements to only a subset of cells at the periphery. To increase mass transfer of viral particles, we designed a microfluidic device that holds islets in parallel hydrodynamic traps connected by an expanding by-pass channel. We modeled viral particle flow into the tissue using fluorescently-labelled gold nanoparticles of varying sizes and showed a penetration threshold of only ∼5 nm. To increase this threshold, we used EDTA to transiently reduce cell-cell adhesion and expand intercellular space. Ultimately, a combination of media flow and ETDA treatment significantly increased adenoviral transduction to the core of the islet. As proof-of-principle, we used this protocol to transduce an ER-targeted redox sensitive sensor (eroGFP), and revealed significantly greater ER redox capacity at core islet cells. Overall, these data demonstrate a robust method to enhance transduction efficiency of islets, and potentially other tissues, by using a combination of microfluidic flow and transient tissue expansion.


Asunto(s)
Adenoviridae/fisiología , Estrés del Retículo Endoplásmico , Técnicas de Transferencia de Gen/instrumentación , Islotes Pancreáticos/virología , Dispositivos Laboratorio en un Chip , Modelos Biológicos , Virión/fisiología , Animales , Quelantes del Calcio/farmacología , Supervivencia Celular/efectos de los fármacos , Ditiotreitol/toxicidad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Diseño de Equipo , Estudios de Factibilidad , Oro/química , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Masculino , Nanopartículas del Metal/química , Ratones Endogámicos C57BL , Prueba de Estudio Conceptual , Proteínas Recombinantes de Fusión/metabolismo , Sustancias Reductoras/toxicidad , Técnicas de Cultivo de Tejidos
16.
PLoS One ; 11(2): e0149177, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26882284

RESUMEN

The Jagn1 protein was indentified in a SILAC proteomic screen of proteins that are increased in insulinoma cells expressing a folding-deficient proinsulin. Jagn1 mRNA was detected in primary rodent islets and in insulinoma cell lines and the levels were increased in response to ER stress. The function of Jagn1 was assessed in insulinoma cells by both knock-down and overexpression approaches. Knock-down of Jagn1 caused an increase in glucose-stimulated insulin secretion resulting from an increase in proinsulin biosynthesis. In contrast, overexpression of Jagn1 in insulinoma cells resulted in reduced cellular proinsulin and insulin levels. Our results identify a novel role for Jagn1 in regulating proinsulin biosynthesis in pancreatic ß-cells. Under ER stress conditions Jagn1 is induced which might contribute to reducing proinsulin biosynthesis, in part by helping to relieve the protein folding load in the ER in an effort to restore ER homeostasis.


Asunto(s)
Estrés del Retículo Endoplásmico , Proteínas de la Membrana/metabolismo , Proinsulina/biosíntesis , Animales , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Insulina/metabolismo , Secreción de Insulina , Insulinoma/metabolismo , Islotes Pancreáticos/metabolismo , Ratones , Proteínas Mutantes/metabolismo , Proteómica , Ratas
17.
PLoS One ; 10(2): e0116410, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25668518

RESUMEN

Reactive oxygen species (ROS) primarily produced via NADPH oxidase play an important role for killing microorganisms in neutrophils. In this study we examined if ROS production in Human promyelocytic leukemia cells (HL60) differentiated into neutrophil-like cells (dHL60) induces ER stress and activates the unfolded protein response (UPR). To cause ROS production cells were treated with PMA or by chronic hyperglycemia. Chronic hyperglycemia failed to induce ROS production and did not cause activation of the UPR in dHL60 cells. PMA, a pharmacologic NADPH oxidase activator, induced ER stress in dHL60 cells as monitored by IRE-1 and PERK pathway activation, and this was independent of calcium signaling. The NADPH oxidase inhibitor, DPI, abolished both ROS production and UPR activation. These results show that ROS produced by NADPH oxidase induces ER stress and suggests a close association between the redox state of the cell and the activation of the UPR in neutrophil-like HL60 cells.


Asunto(s)
Diferenciación Celular/fisiología , Estrés del Retículo Endoplásmico/fisiología , NADPH Oxidasas/metabolismo , Neutrófilos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Respuesta de Proteína Desplegada/fisiología , Análisis de Varianza , Western Blotting , Calcio/metabolismo , Cartilla de ADN/genética , Activación Enzimática/efectos de los fármacos , Citometría de Flujo , Células HL-60 , Humanos , NADPH Oxidasas/antagonistas & inhibidores , Reacción en Cadena en Tiempo Real de la Polimerasa , Acetato de Tetradecanoilforbol/farmacología
18.
Exp Cell Res ; 330(1): 111-22, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25447309

RESUMEN

Endoplasmic reticulum (ER) stress is implicated in pancreatic ß-cell dysfunction and death resulting in type 2 diabetes. Activating transcription factor 6 (ATF6) is an essential component of the Unfolded Protein Response (UPR) and consists of two isoforms, ATF6α and ATF6ß. Here we investigated the role of ATF6ß. ATF6ß mRNA was detected in pancreatic ß-cell lines and rodent and human islets. We also detected ATF6ß protein and production of the active form (ATF6ßp60) in response to ER stress. Knock-down of ATF6ß in INS-1 832/13 insulinoma cells did not affect mRNA induction of several major UPR genes in response to ER stress, suggesting ATF6ß is not essential for the basic UPR. Expressing active ATF6ßp60 or ATF6αp50 followed by microarray analysis showed that they regulate similar UPR genes, although some genes such as Wfs1 are ATF6ß-specific. ATF6ß, but not ATF6α, is able to bind the Wfs1 promoter and induce Wfs1 gene and protein expression. Knock-down of ATF6ß increased the susceptibility of ß-cells to ER stress-induced apoptosis, while overexpression of active ATF6ßp60 reduced apoptosis. Thus, ATF6ß is not essential for induction of most UPR genes, but is required to maintain cell survival in ß-cells undergoing chronic ER stress, which in part relates to its ability to induce Wfs1, a pro-survival gene.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Proteínas de Unión a Calmodulina/metabolismo , Estrés del Retículo Endoplásmico , Células Secretoras de Insulina/metabolismo , Proteínas de la Membrana/metabolismo , Factor de Transcripción Activador 6/genética , Animales , Apoptosis , Proteínas de Unión a Calmodulina/genética , Línea Celular , Supervivencia Celular , Células Cultivadas , Humanos , Proteínas de la Membrana/genética , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Respuesta de Proteína Desplegada
19.
BMC Cell Biol ; 15: 29, 2014 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-25011481

RESUMEN

BACKGROUND: The Akita mutation (C96Y) in the insulin gene results in early onset diabetes in both humans and mice. Expression of mutant proinsulin (C96Y) causes endoplasmic reticulum (ER) stress in pancreatic ß-cells and consequently the cell activates the unfolded protein response (UPR). Since the proinsulin is terminally misfolded ER stress is irremediable and chronic activation of the UPR eventually activates apoptosis in some cells. Here we analyzed the IRE1-dependent activation of genes in response to misfolded proinsulin production in an inducible mutant proinsulin (C96Y) insulinoma cell line. RESULTS: The IRE1 endoribonuclease inhibitors 4µ8c and MKC-3946 prevented the splicing of the XBP1 mRNA in response to ER stress caused by mutant proinsulin production. Microarray expression analysis and qPCR validation of select genes revealed that maximal upregulation of many UPR genes in response to mutant proinsulin production required IRE1, although most were still increased above control. Interestingly, neither degradation of misfolded proinsulin via ER-associated degradation (ERAD), nor apoptosis induced by prolonged misfolded proinsulin expression were affected by inhibiting IRE1. CONCLUSIONS: Although maximal induction of most UPR genes requires IRE1, inhibition of IRE1 does not affect ERAD of misfolded proinsulin or predispose pancreatic ß-cells expressing misfolded proinsulin to chronic ER stress-induced apoptosis.


Asunto(s)
Apoptosis/efectos de los fármacos , Células Secretoras de Insulina/citología , Proteínas de la Membrana/antagonistas & inhibidores , Proinsulina/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Respuesta de Proteína Desplegada/efectos de los fármacos , Animales , Línea Celular Tumoral , Regulación de la Expresión Génica/efectos de los fármacos , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Proteínas de la Membrana/metabolismo , Mutación Puntual , Proinsulina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteolisis/efectos de los fármacos , Ratas
20.
J Cell Sci ; 126(Pt 9): 1962-8, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23444373

RESUMEN

Stromal cell-derived factor 2-like 1 (SDF2L1) is an endoplasmic reticulum (ER)-localized protein whose function is undefined. Here we show that SDF2L1 protein levels are increased in response to ER stress-inducing compounds, but not other cell stressors that we tested in insulinoma cell lines. SDF2L1 protein levels were also induced by expression of misfolded proinsulin in insulinoma cells and in islets from diabetic mice. Immunoprecipitation and binding assays demonstrated that SDF2L1 interacts with the ER chaperone GRP78/BiP, the ER-associated degradation (ERAD) machinery and with misfolded proinsulin. Unexpectedly, knockdown of SDF2L1 in INS-1 (insulin 2 C96Y-GFP) cells increased the degradation kinetics of mutant proinsulin, suggesting that SDF2L1 regulates substrate availability for the ERAD system. We suggest that SDF2L1 increases the time that misfolded proteins have to achieve a correctly folded conformation and thus that SDF2L1 can act as a buffer for substrate availability for ERAD in pancreatic ß-cells.


Asunto(s)
Degradación Asociada con el Retículo Endoplásmico , Retículo Endoplásmico/metabolismo , Células Secretoras de Insulina/metabolismo , Proteínas de la Membrana/metabolismo , Proinsulina/metabolismo , Proteolisis , Animales , Línea Celular Tumoral , Retículo Endoplásmico/genética , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/genética , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Células Secretoras de Insulina/patología , Proteínas de la Membrana/genética , Mutación , Proinsulina/genética , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA