Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 131(17)2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34623328

RESUMEN

Properly balancing microbial responses by the innate immune system through pattern recognition receptors (PRRs) is critical for intestinal immune homeostasis. Ring finger protein 186 (RNF186) genetic variants are associated with inflammatory bowel disease (IBD). However, functions for the E3 ubiquitin ligase RNF186 are incompletely defined. We found that upon stimulation of the PRR nucleotide-binding oligomerization domain containing 2 (NOD2) in human macrophages, RNF186 localized to the ER, formed a complex with ER stress sensors, ubiquitinated the ER stress sensor activating transcription factor 6 (ATF6), and promoted the unfolded protein response (UPR). These events, in turn, led to downstream signaling, cytokine secretion, and antimicrobial pathway induction. Importantly, RNF186-mediated ubiquitination of K152 on ATF6 was required for these outcomes, highlighting a key role for ATF6 ubiquitination in PRR-initiated functions. Human macrophages transfected with the rare RNF186-A64T IBD risk variant and macrophages from common rs6426833 RNF186 IBD risk carriers demonstrated reduced NOD2-induced outcomes, which were restored by rescuing UPR signaling. Mice deficient in RNF186 or ATF6 demonstrated a reduced UPR in colonic tissues, increased weight loss, and less effective clearance of bacteria with dextran sodium sulfate-induced injury and upon oral challenge with Salmonella Typhimurium. Therefore, we identified that RNF186 was required for PRR-induced, UPR-associated signaling leading to key macrophage functions; defined that RNF186-mediated ubiquitination of ATF6 was essential for these functions; and elucidated how RNF186 IBD risk variants modulated these outcomes.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Respuesta de Proteína Desplegada/fisiología , Factor de Transcripción Activador 6/química , Factor de Transcripción Activador 6/deficiencia , Factor de Transcripción Activador 6/genética , Animales , Estrés del Retículo Endoplásmico , Variación Genética , Interacciones Microbiota-Huesped , Humanos , Inmunidad Innata , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Adaptadora de Señalización NOD2/metabolismo , Receptores de Reconocimiento de Patrones/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Factores de Riesgo , Transducción de Señal , Ubiquitina-Proteína Ligasas/deficiencia , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
2.
Circ Res ; 124(1): 79-93, 2019 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-30582446

RESUMEN

RATIONALE: Endoplasmic reticulum (ER) stress dysregulates ER proteostasis, which activates the transcription factor, ATF6 (activating transcription factor 6α), an inducer of genes that enhance protein folding and restore ER proteostasis. Because of increased protein synthesis, it is possible that protein folding and ER proteostasis are challenged during cardiac myocyte growth. However, it is not known whether ATF6 is activated, and if so, what its function is during hypertrophic growth of cardiac myocytes. OBJECTIVE: To examine the activity and function of ATF6 during cardiac hypertrophy. METHODS AND RESULTS: We found that ER stress and ATF6 were activated and ATF6 target genes were induced in mice subjected to an acute model of transverse aortic constriction, or to free-wheel exercise, both of which promote adaptive cardiac myocyte hypertrophy with preserved cardiac function. Cardiac myocyte-specific deletion of Atf6 (ATF6 cKO [conditional knockout]) blunted transverse aortic constriction and exercise-induced cardiac myocyte hypertrophy and impaired cardiac function, demonstrating a role for ATF6 in compensatory myocyte growth. Transcript profiling and chromatin immunoprecipitation identified RHEB (Ras homologue enriched in brain) as an ATF6 target gene in the heart. RHEB is an activator of mTORC1 (mammalian/mechanistic target of rapamycin complex 1), a major inducer of protein synthesis and subsequent cell growth. Both transverse aortic constriction and exercise upregulated RHEB, activated mTORC1, and induced cardiac hypertrophy in wild type mouse hearts but not in ATF6 cKO hearts. Mechanistically, knockdown of ATF6 in neonatal rat ventricular myocytes blocked phenylephrine- and IGF1 (insulin-like growth factor 1)-mediated RHEB induction, mTORC1 activation, and myocyte growth, all of which were restored by ectopic RHEB expression. Moreover, adeno-associated virus 9- RHEB restored cardiac growth to ATF6 cKO mice subjected to transverse aortic constriction. Finally, ATF6 induced RHEB in response to growth factors, but not in response to other activators of ATF6 that do not induce growth, indicating that ATF6 target gene induction is stress specific. CONCLUSIONS: Compensatory cardiac hypertrophy activates ER stress and ATF6, which induces RHEB and activates mTORC1. Thus, ATF6 is a previously unrecognized link between growth stimuli and mTORC1-mediated cardiac growth.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Hipertrofia Ventricular Izquierda/enzimología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Miocitos Cardíacos/enzimología , Proteína Homóloga de Ras Enriquecida en el Cerebro/metabolismo , Activación Transcripcional , Función Ventricular Izquierda , Remodelación Ventricular , Factor de Transcripción Activador 6/deficiencia , Factor de Transcripción Activador 6/genética , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Retículo Endoplásmico/enzimología , Estrés del Retículo Endoplásmico , Predisposición Genética a la Enfermedad , Hipertrofia Ventricular Izquierda/genética , Hipertrofia Ventricular Izquierda/patología , Hipertrofia Ventricular Izquierda/fisiopatología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos Cardíacos/patología , Fenotipo , Pliegue de Proteína , Proteostasis , Proteína Homóloga de Ras Enriquecida en el Cerebro/genética , Transducción de Señal
3.
Matrix Biol ; 70: 50-71, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29522813

RESUMEN

Whilst the role of ATF6α in modulating the unfolded protein response (UPR) has been well documented, the function of its paralogue ATF6ß is less well understood. Using knockdown in cell culture and gene ablation in mice we have directly compared the roles of ATF6α & ß in responding to the increased ER stress induced by mutant forms of type X collagen that cause the ER stress-associated metaphyseal chondrodysplasia type Schmid (MCDS). ATF6α more efficiently deals with the disease-associated ER stress in the absence of ATF6ß and conversely, ATF6ß is less effective in the absence of ATF6α. Furthermore, disease severity in vivo is increased by ATF6α ablation and decreased by ATF6ß ablation. In addition, novel functions for each paralogue are described including an ATF6ß-specific role in controlling growth plate chondrocyte proliferation. The clear demonstration of the intimate relationship of the two ATF6 isoforms and how ATF6ß can moderate the activity of ATF6α and vice versa is of great significance for understanding the UPR mechanism. The activities of both ATF6 isoforms and their separate roles need consideration when deciding how to target increased ER stress as a means of treating MCDS and other ER stress-associated diseases.


Asunto(s)
Factor de Transcripción Activador 6/genética , Condrocitos/metabolismo , Colágeno Tipo X/genética , Placa de Crecimiento/metabolismo , Osteocondrodisplasias/genética , Factor de Transcripción Activador 6/deficiencia , Animales , Proliferación Celular , Condrocitos/patología , Colágeno Tipo X/metabolismo , Modelos Animales de Enfermedad , Estrés del Retículo Endoplásmico , Femenino , Regulación de la Expresión Génica , Placa de Crecimiento/patología , Humanos , Masculino , Ratones , Ratones Noqueados , Mutación , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patología , Cultivo Primario de Células , Índice de Severidad de la Enfermedad , Transducción de Señal , Respuesta de Proteína Desplegada
4.
Glia ; 66(7): 1331-1345, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29436030

RESUMEN

Endoplasmic reticulum (ER) stress and the unfolded protein response (UPR) play a critical role in immune-mediated demyelinating diseases, including multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE), by regulating the viability of oligodendrocytes. Our previous studies show that activation of the PERK branch of the UPR protects myelinating oligodendrocytes against ER stress in young, developing mice that express IFN-γ, a key pro-inflammatory cytokine in MS and EAE, in the CNS. Several studies also demonstrate that PERK activation preserves oligodendrocyte viability and function, protecting mice against EAE. While evidence suggests activation of the ATF6α branch of the UPR in oligodendrocytes under normal and disease conditions, the effects of ATF6α activation on oligodendrocytes in immune-mediated demyelinating diseases remain unknown. Herein, we showed that ATF6α deficiency had no effect on oligodendrocytes under normal conditions. Interestingly, we showed that ATF6α deficiency exacerbated ER stressed-induced myelinating oligodendrocyte death and subsequent myelin loss in the developing CNS of IFN-γ-expressing mice. Moreover, we found that ATF6α deficiency increased EAE severity and aggravated EAE-induced oligodendrocyte loss and demyelination, without affecting inflammation. Thus, these data suggest the protective effects of ATF6α activation on oligodendrocytes in immune-mediated demyelinating diseases.


Asunto(s)
Factor de Transcripción Activador 6/deficiencia , Muerte Celular/fisiología , Encefalomielitis Autoinmune Experimental/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Oligodendroglía/metabolismo , Factor de Transcripción Activador 6/genética , Animales , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Encéfalo/patología , Supervivencia Celular/fisiología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Interferón gamma/genética , Interferón gamma/metabolismo , Leucocitos/metabolismo , Leucocitos/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Glicoproteína Mielina-Oligodendrócito , Oligodendroglía/patología , Fragmentos de Péptidos , Médula Espinal/crecimiento & desarrollo , Médula Espinal/metabolismo , Médula Espinal/patología , Bazo/metabolismo , Bazo/patología
5.
Circ Res ; 120(5): 862-875, 2017 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-27932512

RESUMEN

RATIONALE: Endoplasmic reticulum (ER) stress causes the accumulation of misfolded proteins in the ER, activating the transcription factor, ATF6 (activating transcription factor 6 alpha), which induces ER stress response genes. Myocardial ischemia induces the ER stress response; however, neither the function of this response nor whether it is mediated by ATF6 is known. OBJECTIVE: Here, we examined the effects of blocking the ATF6-mediated ER stress response on ischemia/reperfusion (I/R) in cardiac myocytes and mouse hearts. METHODS AND RESULTS: Knockdown of ATF6 in cardiac myocytes subjected to I/R increased reactive oxygen species and necrotic cell death, both of which were mitigated by ATF6 overexpression. Under nonstressed conditions, wild-type and ATF6 knockout mouse hearts were similar. However, compared with wild-type, ATF6 knockout hearts showed increased damage and decreased function after I/R. Mechanistically, gene array analysis showed that ATF6, which is known to induce genes encoding ER proteins that augment ER protein folding, induced numerous oxidative stress response genes not previously known to be ATF6-inducible. Many of the proteins encoded by the ATF6-induced oxidative stress genes identified here reside outside the ER, including catalase, which is known to decrease damaging reactive oxygen species in the heart. Catalase was induced by the canonical ER stressor, tunicamycin, and by I/R in cardiac myocytes from wild-type but not in cardiac myocytes from ATF6 knockout mice. ER stress response elements were identified in the catalase gene and were shown to bind ATF6 in cardiac myocytes, which increased catalase promoter activity. Overexpression of catalase, in vivo, restored ATF6 knockout mouse heart function to wild-type levels in a mouse model of I/R, as did adeno-associated virus 9-mediated ATF6 overexpression. CONCLUSIONS: ATF6 serves an important role as a previously unappreciated link between the ER stress and oxidative stress gene programs, supporting a novel mechanism by which ATF6 decreases myocardial I/R damage.


Asunto(s)
Factor de Transcripción Activador 6/biosíntesis , Estrés del Retículo Endoplásmico/fisiología , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Estrés Oxidativo/fisiología , Factor de Transcripción Activador 6/deficiencia , Animales , Animales Recién Nacidos , Células HEK293 , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/patología , Miocitos Cardíacos , Ratas , Ratas Sprague-Dawley , Transducción de Señal/fisiología
6.
J Neurochem ; 139(6): 1124-1137, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27333444

RESUMEN

Accumulating evidence suggests a critical role for the unfolded protein response in multiple sclerosis (MS) and in its animal model, experimental autoimmune encephalomyelitis (EAE). In this study, we investigated the relevance of activating transcription factor 6α (ATF6α), an upstream regulator of part of the unfolded protein response, in EAE. The expressions of ATF6α-target molecular chaperones such as glucose-regulated protein 78 (GRP78) and glucose-regulated protein 94 (GRP94) were enhanced in the acute inflammatory phase after induction of EAE. Deletion of Atf6α suppressed the accumulation of T cells and microglia/macrophages in the spinal cord, and ameliorated the clinical course and demyelination after EAE induction. In contrast to the phenotypes in the spinal cord, activation status of T cells in the peripheral tissues or in the culture system was not different between two genotypes. Bone marrow transfer experiments and adoptive transfer of autoimmune CD4+ T cells to recipient mice (passive EAE) also revealed that CNS-resident cells are responsible for the phenotypes observed in Atf6α-/- mice. Further experiments with cultured cells indicated that inflammatory response was reduced in Atf6α-/- microglia, but not in Atf6α-/- astrocytes, and was associated with proteasome-dependent degradation of NF-κB p65. Thus, our results demonstrate a novel role for ATF6α in microglia-mediated CNS inflammation. We investigated the relevance of ATF6α, an upstream regulator of part of the UPR, in EAE. Deletion of Atf6α suppressed inflammation, and ameliorated demyelination after EAE. Bone marrow transfer experiments and adoptive transfer of autoimmune CD4+ T cells revealed that CNS-resident cells are responsible for the phenotypes in Atf6α-/- mice. Furthermore, inflammatory response was reduced in Atf6α-/- microglia, and was associated with degradation of NF-κB p65. Our results demonstrate a novel role for ATF6α in microglia-mediated inflammation. Cover image for this issue: doi: 10.1111/jnc.13346.


Asunto(s)
Factor de Transcripción Activador 6/deficiencia , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Microglía/metabolismo , Animales , Células Cultivadas , Encefalomielitis Autoinmune Experimental/prevención & control , Chaperón BiP del Retículo Endoplásmico , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
7.
Nat Commun ; 6: 6496, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25754093

RESUMEN

Endoplasmic reticulum (ER) stress is associated with diabetic nephropathy (DN), but its pathophysiological relevance and the mechanisms that compromise adaptive ER signalling in podocytes remain unknown. Here we show that nuclear translocation of the transcription factor spliced X-box binding protein-1 (sXBP1) is selectively impaired in DN, inducing activating transcription factor-6 (ATF6) and C/EBP homology protein (CHOP). Podocyte-specific genetic ablation of XBP1 or inducible expression of ATF6 in mice aggravates DN. sXBP1 lies downstream of insulin signalling and attenuating podocyte insulin signalling by genetic ablation of the insulin receptor or the regulatory subunits phosphatidylinositol 3-kinase (PI3K) p85α or p85ß impairs sXBP1 nuclear translocation and exacerbates DN. Corroborating our findings from murine DN, the interaction of sXBP1 with p85α and p85ß is markedly impaired in the glomerular compartment of human DN. Thus, signalling via the insulin receptor, p85, and XBP1 maintains podocyte homeostasis, while disruption of this pathway impairs podocyte function in DN.


Asunto(s)
Factor de Transcripción Activador 6/genética , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Proteínas de Unión al ADN/genética , Diabetes Mellitus Experimental/genética , Nefropatías Diabéticas/genética , Estrés del Retículo Endoplásmico/genética , Podocitos/metabolismo , Factores de Transcripción/genética , Factor de Transcripción Activador 6/deficiencia , Animales , Fosfatidilinositol 3-Quinasa Clase Ia/deficiencia , Proteínas de Unión al ADN/deficiencia , Bases de Datos Factuales , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/inducido químicamente , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/patología , Regulación de la Expresión Génica , Humanos , Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Podocitos/patología , Receptor de Insulina/deficiencia , Receptor de Insulina/genética , Factores de Transcripción del Factor Regulador X , Transducción de Señal , Estreptozocina , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Factores de Transcripción/deficiencia , Proteína 1 de Unión a la X-Box
8.
Respir Res ; 14: 141, 2013 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-24364984

RESUMEN

BACKGROUND: The endoplasmic reticulum (ER) stress response participates in many chronic inflammatory and autoimmune diseases. In the current study, we sought to examine the contribution of ER stress transducers in the pathogenesis of three principal facets of allergic asthma: inflammation, airway fibrosis, and airways hyperresponsiveness. METHODS: House Dust Mite (HDM) was used as an allergen for in vitro and in vivo challenge of primary human and murine airway epithelial cells. ER stress transducers were modulated using specific small interfering RNAs (siRNAs) in vivo. Inflammation, airway remodeling, and hyperresponsiveness were measured by total bronchoalveolar lavage (BAL) cell counts, determination of collagen, and methacholine responsiveness in mice, respectively. RESULTS: Challenge of human bronchiolar and nasal epithelial cells with HDM extract induced the ER stress transducer, activating transcription factor 6 α (ATF6α) as well as protein disulfide isomerase, ERp57, in association with activation of caspase-3. SiRNA-mediated knockdown of ATF6α and ERp57 during HDM administration in mice resulted in a decrease in components of HDM-induced ER stress, disulfide mediated oligomerization of Bak, and activation of caspase-3. Furthermore, siRNA-mediated knockdown of ATF6α and ERp57 led to decreased inflammation, airway hyperresponsiveness and airway fibrosis. CONCLUSION: Collectively, our work indicates that HDM induces ER stress in airway epithelial cells and that ATF6α and ERp57 play a significant role in the development of cardinal features of allergic airways disease. Inhibition of ER stress responses may provide a potential therapeutic avenue in chronic asthma and sub-epithelial fibrosis associated with loss of lung function.


Asunto(s)
Apoptosis , Bronquios/patología , Estrés del Retículo Endoplásmico/fisiología , Células Epiteliales/patología , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/fisiopatología , Pyroglyphidae/fisiología , Factor de Transcripción Activador 6/deficiencia , Factor de Transcripción Activador 6/efectos de los fármacos , Factor de Transcripción Activador 6/genética , Animales , Bronquios/metabolismo , Bronquios/fisiopatología , Caspasa 3/metabolismo , Línea Celular , Células Cultivadas , Colágeno/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Humanos , Técnicas In Vitro , Cloruro de Metacolina/metabolismo , Ratones , Ratones Endogámicos BALB C , Proteína Disulfuro Isomerasas/deficiencia , Proteína Disulfuro Isomerasas/efectos de los fármacos , Proteína Disulfuro Isomerasas/genética , Fibrosis Pulmonar/metabolismo , ARN Interferente Pequeño/farmacología
9.
Metabolism ; 61(8): 1118-28, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22386934

RESUMEN

Activating transcription factor 6α (ATF6α) is essential for the endoplasmic reticulum (ER) stress response. Since recent studies suggested that ER stress is involved in the pathogenesis of type 2 diabetes mellitus, we have analyzed Atf6α-null (Atf6α(-/-)) mice challenged with metabolic overload or genetic manipulations. Atf6α(-/-) mice were fed a high-fat diet to create diet-induced obese (DO) mice, and were subjected to examination of glucose homeostasis with biochemical and morphological analysis of the pancreatic ß-cell and liver tissues. Atf6α-null mice were also crossed with genetic models of diabetes caused either by insulin resistance (Agouti obese mice) or by impaired insulin secretion (Ins2(WT/C96Y) mice). Atf6α(-/-) DO mice were less glucose tolerant with blunted insulin secretion compared to littermates on a high-fat diet. Pancreatic insulin content was lower in Atf6α(-/-) DO mice with the swollen ß-cell ER, a typical feature of cells with ER stress. In the liver of Atf6α(-/-) DO mice, XBP-1 splicing was increased, suggesting that higher ER stress was present. ATF6-deficient mice showed increased mRNA expressions of glucose-6-phosphatase and SREBP1c associated with a tendency for a higher degree of steatosis in the liver. However, Atf6α(-/-) DO mice exhibited higher insulin sensitivity with lower serum triglyceride levels. Similar phenotypes were observed in ATF6α-deficient Agouti mice. In addition, ATF6α-deficiency accelerated reduction in pancreatic insulin content in Ins2(WT/C96Y) mice. These data suggested that ATF6α contributes to both prevention and promotion of diabetes; it protects ß-cells from ER stress and suppresses hepatosteatosis, but plays a role in the development of hyperlipidemia and insulin resistance.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Glucemia/metabolismo , Diabetes Mellitus/metabolismo , Dieta Alta en Grasa , Retículo Endoplásmico/metabolismo , Hígado Graso/metabolismo , Hiperlipidemias/metabolismo , Resistencia a la Insulina , Células Secretoras de Insulina/metabolismo , Obesidad/metabolismo , Factor de Transcripción Activador 6/deficiencia , Factor de Transcripción Activador 6/genética , Animales , Diabetes Mellitus/etiología , Diabetes Mellitus/patología , Diabetes Mellitus/prevención & control , Dieta Alta en Grasa/efectos adversos , Femenino , Glucosa-6-Fosfatasa/genética , Glucosa-6-Fosfatasa/metabolismo , Homeostasis , Hiperlipidemias/sangre , Células Secretoras de Insulina/ultraestructura , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Obesidad/etiología , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Triglicéridos/sangre
10.
J Exp Med ; 208(7): 1533-46, 2011 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-21670204

RESUMEN

The ROP18 kinase has been identified as a key virulence determinant conferring a high mortality phenotype characteristic of type I Toxoplasma gondii strains. This major effector molecule is secreted by the rhoptries into the host cells during invasion; however, the molecular mechanisms by which this kinase exerts its pathogenic action remain poorly understood. In this study, we show that ROP18 targets the host endoplasmic reticulum-bound transcription factor ATF6ß. Disruption of the ROP18 gene severely impairs acute toxoplasmosis by the type I RH strain. Because another virulence factor ROP16 kinase modulates immune responses through its N-terminal portion, we focus on the role of the N terminus of ROP18 in the subversion of host cellular functions. The N-terminal extension of ROP18 contributes to ATF6ß-dependent pathogenicity by interacting with ATF6ß and destabilizing it. The kinase activity of ROP18 is essential for proteasome-dependent degradation of ATF6ß and for parasite virulence. Consistent with a key role for ATF6ß in resistance against this intracellular pathogen, ATF6ß-deficient mice exhibit a high susceptibility to infection by ROP18-deficient parasites. The results reveal that interference with ATF6ß-dependent immune responses is a novel pathogenic mechanism induced by ROP18.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Proteínas Serina-Treonina Quinasas/toxicidad , Toxoplasma/patogenicidad , Factores de Virulencia/toxicidad , Factor de Transcripción Activador 6/deficiencia , Factor de Transcripción Activador 6/genética , Factor de Transcripción Activador 6/inmunología , Animales , Animales Modificados Genéticamente , Técnicas de Inactivación de Genes , Genes Protozoarios , Interacciones Huésped-Parásitos/inmunología , Interacciones Huésped-Parásitos/fisiología , Hipersensibilidad Inmediata/etiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Ratones Noqueados , Proteínas Serina-Treonina Quinasas/genética , Proteínas Protozoarias , Toxoplasma/enzimología , Toxoplasma/genética , Toxoplasmosis Animal/etiología , Toxoplasmosis Animal/inmunología , Virulencia/fisiología , Factores de Virulencia/genética
11.
Mol Biol Cell ; 21(17): 2975-86, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20631254

RESUMEN

Accumulation of unfolded proteins in the endoplasmic reticulum (ER) activates homeostatic responses collectively termed the unfolded protein response. Among the three principal signaling pathways operating in mammals, activating transcription factor (ATF)6alpha plays a pivotal role in transcriptional induction of ER-localized molecular chaperones and folding enzymes as well as components of ER-associated degradation, and thereby mouse embryonic fibroblasts deficient in ATF6alpha are sensitive to ER stress. However, ATF6alpha-knockout mice show no apparent phenotype under normal growing conditions. In this report, we burdened mice with intraperitoneal injection of the ER stress-inducing reagent tunicamycin and found that wild-type mice were able to recover from the insult, whereas ATF6alpha-knockout mice exhibited liver dysfunction and steatosis. Thus, ATF6alpha-knockout mice accumulated neutral lipids in the liver such as triacylglycerol and cholesterol, which was ascribable to blockage of beta-oxidation of fatty acids caused by decreased mRNA levels of the enzymes involved in the process, suppression of very-low-density lipoprotein formation due to destabilized apolipoprotein B-100, and stimulation of lipid droplet formation resulting from transcriptional induction of adipose differentiation-related protein. Accordingly, the hepatocytes of tunicamycin-injected knockout mice were filled with many lipid droplets. These results establish links among ER stress, lipid metabolism, and steatosis.


Asunto(s)
Factor de Transcripción Activador 6/deficiencia , Retículo Endoplásmico/patología , Hígado Graso/genética , Metabolismo de los Lípidos/genética , Estrés Fisiológico/efectos de los fármacos , Tunicamicina/farmacología , Factor de Transcripción Activador 6/genética , Factor de Transcripción Activador 6/metabolismo , Animales , Apolipoproteína B-100/metabolismo , Peso Corporal/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Hígado Graso/patología , Hígado Graso/fisiopatología , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Células Hep G2 , Humanos , Inyecciones Intraperitoneales , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/patología , Hígado/fisiopatología , Hígado/ultraestructura , Pruebas de Función Hepática , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Estrés Fisiológico/inmunología , Tunicamicina/administración & dosificación
12.
Dev Cell ; 13(3): 322-4, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17765676
13.
Dev Cell ; 13(3): 351-64, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17765679

RESUMEN

In vertebrates, three proteins--PERK, IRE1alpha, and ATF6alpha--sense protein-misfolding stress in the ER and initiate ER-to-nucleus signaling cascades to improve cellular function. The mechanism by which this unfolded protein response (UPR) protects ER function during stress is not clear. To address this issue, we have deleted Atf6alpha in the mouse. ATF6alpha is neither essential for basal expression of ER protein chaperones nor for embryonic or postnatal development. However, ATF6alpha is required in both cells and tissues to optimize protein folding, secretion, and degradation during ER stress and thus to facilitate recovery from acute stress and tolerance to chronic stress. Challenge of Atf6alpha null animals in vivo compromises organ function and survival despite functional overlap between UPR sensors. These results suggest that the vertebrate ATF6alpha pathway evolved to maintain ER function when cells are challenged with chronic stress and provide a rationale for the overlap among the three UPR pathways.


Asunto(s)
Factor de Transcripción Activador 6/deficiencia , Factor de Transcripción Activador 6/metabolismo , Retículo Endoplásmico/metabolismo , Estrés Oxidativo , Factor de Transcripción Activador 6/genética , Alelos , Animales , Células Cultivadas , Enfermedad Crónica , Cruzamientos Genéticos , Ditioeritritol/farmacología , Exones , Fibroblastos/metabolismo , Eliminación de Gen , Perfilación de la Expresión Génica , Integrasas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Pliegue de Proteína , ARN Mensajero/metabolismo , Reactivos de Sulfhidrilo/farmacología , Transactivadores/genética , Transactivadores/metabolismo , Tunicamicina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...