Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros











Intervalo de año de publicación
1.
Immunity ; 57(6): 1225-1242.e6, 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38749446

RESUMEN

Classical monocytes (CMs) are ephemeral myeloid immune cells that circulate in the blood. Emerging evidence suggests that CMs can have distinct ontogeny and originate from either granulocyte-monocyte- or monocyte-dendritic-cell progenitors (GMPs or MDPs). Here, we report surface markers that allowed segregation of murine GMP- and MDP-derived CMs, i.e., GMP-Mo and MDP-Mo, as well as their functional characterization, including fate definition following adoptive cell transfer. GMP-Mo and MDP-Mo yielded an equal increase in homeostatic CM progeny, such as blood-resident non-classical monocytes and gut macrophages; however, these cells differentially seeded various other selected tissues, including the dura mater and lung. Specifically, GMP-Mo and MDP-Mo differentiated into distinct interstitial lung macrophages, linking CM dichotomy to previously reported pulmonary macrophage heterogeneity. Collectively, we provide evidence for the existence of two functionally distinct CM subsets in the mouse that differentially contribute to peripheral tissue macrophage populations in homeostasis and following challenge.


Asunto(s)
Diferenciación Celular , Macrófagos , Monocitos , Animales , Monocitos/inmunología , Monocitos/citología , Ratones , Diferenciación Celular/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Pulmón/citología , Pulmón/inmunología , Homeostasis , Ratones Endogámicos C57BL , Células Dendríticas/inmunología , Linaje de la Célula , Traslado Adoptivo
2.
Methods Mol Biol ; 2713: 253-267, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37639128

RESUMEN

Global gene expression profiling has provided valuable insights into the specific contributions of different cell types to various physiological processes. Notably though, both bulk and single-cell transcriptomics require the prior retrieval of the cells from their tissue context to be analyzed. Isolation protocols for tissue macrophages are, however, notoriously inefficient and, moreover, prone to introduce considerable bias and artifacts. Here, we will discuss a valuable alternative, originally introduced by Amieux and colleagues. This so-called RiboTag approach allows, in combination with respective macrophage-specific Cre transgenic lines, to retrieve macrophage translatomes from crude tissue extracts. We will review our experience with this ingenious method, focusing on the study of brain macrophages, including microglia and border-associated cells. We will elaborate on the advantages of the RiboTag approach that render it a valuable complement to standard cell sorting-based profiling strategies, especially for the investigation of tissue macrophages.


Asunto(s)
Artefactos , Macrófagos , Animales , Animales Modificados Genéticamente , Encéfalo , Separación Celular
3.
Methods Mol Biol ; 2713: 481-503, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37639143

RESUMEN

Macrophages represent a broad spectrum of distinct, but closely related tissue-resident immune cells. This presents a major challenge for the study of functional aspects of these cells using classical Cre recombinase-mediated conditional mutagenesis in mice, since single promoter-driven Cre transgenic models often display limited specificity toward their intended target. The advent of CRISPR/Cas9 technology has now provided a time- and cost-effective method to explore the full potential of binary transgenic, intersectional genetics. Specifically, the use of two promoters driving inactive Cre fragments that, when co-expressed, dimerize and only then gain recombinase activity allows the characterization and manipulation of genetically defined tissue macrophage subpopulations. Here, we will elaborate on the use of this protocol to capitalize on these recent technological advances in mouse genetics and discuss their strengths and pitfalls to improve the study of tissue macrophage subpopulations in physiology and pathophysiology.


Asunto(s)
Técnicas de Transferencia de Gen , Macrófagos , Animales , Ratones , Animales Modificados Genéticamente , Dimerización , Mutagénesis
5.
Immunity ; 54(1): 176-190.e7, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33333014

RESUMEN

The developmental and molecular heterogeneity of tissue macrophages is unravelling, as are their diverse contributions to physiology and pathophysiology. Moreover, also given tissues harbor macrophages in discrete anatomic locations. Functional contributions of specific cell populations can in mice be dissected using Cre recombinase-mediated mutagenesis. However, single promoter-based Cre models show limited specificity for cell types. Focusing on macrophages in the brain, we establish here a binary transgenic system involving complementation-competent NCre and CCre fragments whose expression is driven by distinct promoters: Sall1ncre: Cx3cr1ccre mice specifically target parenchymal microglia and compound transgenic Lyve1ncre: Cx3cr1ccre animals target vasculature-associated macrophages, in the brain, as well as other tissues. We imaged the respective cell populations and retrieved their specific translatomes using the RiboTag in order to define them and analyze their differential responses to a challenge. Collectively, we establish the value of binary transgenesis to dissect tissue macrophage compartments and their functions.


Asunto(s)
Encéfalo/citología , Sistema Nervioso Central/fisiología , Integrasas/metabolismo , Macrófagos/fisiología , Microglía/fisiología , Animales , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Especificidad de Órganos
6.
Immunity ; 53(5): 1033-1049.e7, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33049219

RESUMEN

Microglia, the resident macrophages of the brain parenchyma, are key players in central nervous system (CNS) development, homeostasis, and disorders. Distinct brain pathologies seem associated with discrete microglia activation modules. How microglia regain quiescence following challenges remains less understood. Here, we explored the role of the interleukin-10 (IL-10) axis in restoring murine microglia homeostasis following a peripheral endotoxin challenge. Specifically, we show that lipopolysaccharide (LPS)-challenged mice harboring IL-10 receptor-deficient microglia displayed neuronal impairment and succumbed to fatal sickness. Addition of a microglial tumor necrosis factor (TNF) deficiency rescued these animals, suggesting a microglia-based circuit driving pathology. Single cell transcriptome analysis revealed various IL-10 producing immune cells in the CNS, including most prominently Ly49D+ NK cells and neutrophils, but not microglia. Collectively, we define kinetics of the microglia response to peripheral endotoxin challenge, including their activation and robust silencing, and highlight the critical role of non-microglial IL-10 in preventing deleterious microglia hyperactivation.


Asunto(s)
Endotoxinas/inmunología , Interleucina-10/metabolismo , Microglía/inmunología , Microglía/metabolismo , Animales , Biomarcadores , Encéfalo/inmunología , Encéfalo/metabolismo , Encéfalo/patología , Células Cultivadas , Inmunofenotipificación , Interleucina-10/genética , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Lipopolisacáridos/inmunología , Activación de Macrófagos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones
7.
Elife ; 92020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31916932

RESUMEN

Monocytes are circulating short-lived macrophage precursors that are recruited on demand from the blood to sites of inflammation and challenge. In steady state, classical monocytes give rise to vasculature-resident cells that patrol the luminal side of the endothelium. In addition, classical monocytes feed macrophage compartments of selected organs, including barrier tissues, such as the skin and intestine, as well as the heart. Monocyte differentiation under conditions of inflammation has been studied in considerable detail. In contrast, monocyte differentiation under non-inflammatory conditions remains less well understood. Here we took advantage of a combination of cell ablation and precursor engraftment to investigate the generation of gut macrophages from monocytes. Collectively, we identify factors associated with the gradual adaptation of monocytes to tissue residency. Moreover, comparison of monocyte differentiation into the colon and ileum-resident macrophages revealed the graduated acquisition of gut segment-specific gene expression signatures.


Asunto(s)
Diferenciación Celular , Colon/fisiología , Íleon/fisiología , Macrófagos/metabolismo , Monocitos/citología , Animales , Ratones , Organismos Libres de Patógenos Específicos
8.
Eur J Immunol ; 50(3): 353-362, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31762013

RESUMEN

Conditional mutagenesis and fate mapping have contributed considerably to our understanding of physiology and pathology. Specifically, Cre recombinase-based approaches allow the definition of cell type-specific contributions to disease development and of inter-cellular communication circuits in respective animal models. Here we compared Cx3 cr1CreER and Sall1CreER transgenic mice and their use to decipher the brain macrophage compartment as a showcase to discuss recent technological advances. Specifically, we highlight the need to define the accuracy of Cre recombinase expression, as well as strengths and pitfalls of these particular systems that should be taken into consideration when applying these models.


Asunto(s)
Encéfalo , Integrasas , Macrófagos , Ratones Transgénicos , Modelos Animales , Animales , Ratones , Factores de Transcripción
9.
Eur J Immunol ; 49(1): 19-29, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30407631

RESUMEN

The involvement of macrophages in the pathogenesis of obesity has been recognized since 2003. Early studies mostly focused on the role of macrophages in adipose tissue (AT) and in obesity-associated chronic low-grade inflammation. Lately, AT macrophages were shown to undergo intrinsic metabolic changes that affect their immune function (i.e., immunometabolism), corresponding to their unique properties along the range of pro- versus anti-inflammatory activity. In parallel, recent studies in mice revealed critical neuronal-macrophage interactions, both in the CNS and in peripheral tissues, including in white and brown AT. These intercellular activities impinge on energy and metabolic homeostasis, partially by also engaging adipocytes in a neuronal-macrophage-adipocyte ménage à trois. Finally, neuropeptides (NP), such as NPY and appetite-reducing NPFF, may prove as mediators in such intercellular network. In this concise review, we highlight some of these recent insights on adipose macrophage immunometabolism, as well as central and peripheral neuronal-macrophage interactions with emphasis on their impact on adipocyte biology and whole-body metabolism. We also discuss the expanding view on the role of the NP, NPY and NPFF, in obesity.


Asunto(s)
Tejido Adiposo/fisiología , Inflamación/inmunología , Macrófagos/fisiología , Neuronas/fisiología , Obesidad/inmunología , Animales , Comunicación Celular , Sistema Nervioso Central , Humanos , Neuropéptidos/metabolismo
10.
Nat Immunol ; 18(6): 665-674, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28459435

RESUMEN

Tissue macrophages provide immunological defense and contribute to the establishment and maintenance of tissue homeostasis. Here we used constitutive and inducible mutagenesis to delete the nuclear transcription regulator Mecp2 in macrophages. Mice that lacked the gene encoding Mecp2, which is associated with Rett syndrome, in macrophages did not show signs of neurodevelopmental disorder but displayed spontaneous obesity, which was linked to impaired function of brown adipose tissue (BAT). Specifically, mutagenesis of a BAT-resident Cx3Cr1+ macrophage subpopulation compromised homeostatic thermogenesis but not acute, cold-induced thermogenesis. Mechanistically, malfunction of BAT in pre-obese mice with mutant macrophages was associated with diminished sympathetic innervation and local titers of norepinephrine, which resulted in lower expression of thermogenic factors by adipocytes. Mutant macrophages overexpressed the signaling receptor and ligand PlexinA4, which might contribute to the phenotype by repulsion of sympathetic axons expressing the transmembrane semaphorin Sema6A. Collectively, we report a previously unappreciated homeostatic role for macrophages in the control of tissue innervation. Disruption of this circuit in BAT resulted in metabolic imbalance.


Asunto(s)
Tejido Adiposo Pardo/inmunología , Macrófagos/inmunología , Proteína 2 de Unión a Metil-CpG/genética , Sistema Nervioso Simpático/metabolismo , Termogénesis/inmunología , Adipocitos Marrones , Tejido Adiposo Pardo/inervación , Tejido Adiposo Pardo/metabolismo , Animales , Axones/metabolismo , Receptor 1 de Quimiocinas CX3C , Metabolismo Energético/inmunología , Citometría de Flujo , Homeostasis , Immunoblotting , Macrófagos/metabolismo , Ratones , Mutagénesis Sitio-Dirigida , Proteínas del Tejido Nervioso/metabolismo , Norepinefrina/metabolismo , Obesidad/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Superficie Celular/metabolismo , Receptores de Quimiocina/metabolismo , Semaforinas/metabolismo
11.
Eur J Immunol ; 47(7): 1142-1152, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28471480

RESUMEN

T-cell development is a spatially and temporally regulated process, orchestrated by well-defined contributions of transcription factors and cytokines. Here, we identify the noncoding RNA miR-142 as an additional regulatory layer within murine thymocyte development and proliferation. MiR-142 deficiency impairs the expression of cell cycle-promoting genes in mature mouse thymocytes and early progenitors, accompanied with increased levels of cyclin-dependent kinase inhibitor 1B (Cdkn1b, also known as p27Kip1 ). By using CRISPR/Cas9 technology to delete the miR-142-3p recognition element in the 3'UTR of cdkn1b, we confirm that this gene is a novel target of miR-142-3p in vivo. Increased Cdkn1b protein expression alone however was insufficient to cause proliferation defects in thymocytes, indicating the existence of additional critical miR-142 targets. Collectively, we establish a key role for miR-142 in the control of early and mature thymocyte proliferation, demonstrating the multifaceted role of a single miRNA on several target genes.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , MicroARNs/metabolismo , Timocitos/fisiología , Regiones no Traducidas 3' , Animales , Sistemas CRISPR-Cas , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación Neoplásica de la Expresión Génica , Ratones , MicroARNs/genética , Procesamiento Postranscripcional del ARN
12.
Cell Death Differ ; 24(1): 132-143, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27740623

RESUMEN

Earlier reported small interfering RNA (siRNA) high-throughput screens, identified seven-transmembrane superfamily member 3 (TM7SF3) as a novel inhibitor of pancreatic ß-cell death. Here we show that TM7SF3 maintains protein homeostasis and promotes cell survival through attenuation of ER stress. Overexpression of TM7SF3 inhibits caspase 3/7 activation. In contrast, siRNA-mediated silencing of TM7SF3 accelerates ER stress and activation of the unfolded protein response (UPR). This involves inhibitory phosphorylation of eukaryotic translation initiation factor 2α activity and increased expression of activating transcription factor-3 (ATF3), ATF4 and C/EBP homologous protein, followed by induction of apoptosis. This process is observed both in human pancreatic islets and in a number of cell lines. Some of the effects of TM7SF3 silencing are evident both under basal conditions, in otherwise untreated cells, as well as under different stress conditions induced by thapsigargin, tunicamycin or a mixture of pro-inflammatory cytokines (tumor necrosis factor alpha, interleukin-1 beta and interferon gamma). Notably, TM7SF3 is a downstream target of p53: activation of p53 by Nutlin increases TM7SF3 expression in a time-dependent manner, although silencing of p53 abrogates this effect. Furthermore, p53 is found in physical association with the TM7SF3 promoter. Interestingly, silencing of TM7SF3 promotes p53 activity, suggesting the existence of a negative-feedback loop, whereby p53 promotes expression of TM7SF3 that acts to restrict p53 activity. Our findings implicate TM7SF3 as a novel p53-regulated pro-survival homeostatic factor that attenuates the development of cellular stress and the subsequent induction of the UPR.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Factor de Transcripción Activador 3/metabolismo , Factor de Transcripción Activador 4/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células HEK293 , Células Hep G2 , Humanos , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Ratones , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Tapsigargina/toxicidad , Factor de Transcripción CHOP/metabolismo , Tunicamicina/toxicidad , Respuesta de Proteína Desplegada/efectos de los fármacos , eIF-2 Quinasa/antagonistas & inhibidores , eIF-2 Quinasa/metabolismo
13.
Elife ; 4: e05914, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25955862

RESUMEN

Skeletal integrity is maintained by the co-ordinated activity of osteoblasts, the bone-forming cells, and osteoclasts, the bone-resorbing cells. In this study, we show that mice overexpressing galectin-8, a secreted mammalian lectin of the galectins family, exhibit accelerated osteoclasts activity and bone turnover, which culminates in reduced bone mass, similar to cases of postmenopausal osteoporosis and cancerous osteolysis. This phenotype can be attributed to a direct action of galectin-8 on primary cultures of osteoblasts that secrete the osteoclastogenic factor RANKL upon binding of galectin-8. This results in enhanced differentiation into osteoclasts of the bone marrow cells co-cultured with galectin-8-treated osteoblasts. Secretion of RANKL by galectin-8-treated osteoblasts can be attributed to binding of galectin-8 to receptor complexes that positively (uPAR and MRC2) and negatively (LRP1) regulate galectin-8 function. Our findings identify galectins as new players in osteoclastogenesis and bone remodeling, and highlight a potential regulation of bone mass by animal lectins.


Asunto(s)
Células de la Médula Ósea/metabolismo , Resorción Ósea/genética , Huesos/metabolismo , Galectinas/genética , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Ligando RANK/genética , Animales , Densidad Ósea/genética , Células de la Médula Ósea/citología , Resorción Ósea/metabolismo , Resorción Ósea/patología , Huesos/citología , Técnicas de Cocultivo , Femenino , Galectinas/metabolismo , Regulación de la Expresión Génica , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Transgénicos , Osteoblastos/citología , Osteoclastos/citología , Osteogénesis/genética , Cultivo Primario de Células , Unión Proteica , Ligando RANK/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Receptores de LDL/genética , Receptores de LDL/metabolismo , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
14.
Hepatology ; 57(2): 525-32, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22911490

RESUMEN

UNLABELLED: Sphingolipids are important structural components of cell membranes and act as critical regulators of cell function by modulating intracellular signaling pathways. Specific sphingolipids, such as ceramide, glucosylceramide, and ganglioside GM3, have been implicated in various aspects of insulin resistance, because they have been shown to modify several steps in the insulin signaling pathway, such as phosphorylation of either protein kinase B (Akt) or of the insulin receptor. We now explore the role of the ceramide acyl chain length in insulin signaling by using a ceramide synthase 2 (CerS2) null mouse, which is unable to synthesize very long acyl chain (C22-C24) ceramides. CerS2 null mice exhibited glucose intolerance despite normal insulin secretion from the pancreas. Both insulin receptor and Akt phosphorylation were abrogated in liver, but not in adipose tissue or in skeletal muscle. The lack of insulin receptor phosphorylation in liver correlated with its inability to translocate into detergent-resistant membranes (DRMs). Moreover, DRMs in CerS2 null mice displayed properties significantly different from those in wild-type mice, suggesting that the altered sphingolipid acyl chain length directly affects insulin receptor translocation and subsequent signaling. CONCLUSION: We conclude that the sphingolipid acyl chain composition of liver regulates insulin signaling by modifying insulin receptor translocation into membrane microdomains.


Asunto(s)
Intolerancia a la Glucosa/etiología , Resistencia a la Insulina , Microdominios de Membrana/efectos de los fármacos , Esfingolípidos/metabolismo , Animales , Glucemia/metabolismo , Membrana Celular/efectos de los fármacos , Ceramidas/metabolismo , Intolerancia a la Glucosa/sangre , Insulina/fisiología , Hígado/metabolismo , Microdominios de Membrana/fisiología , Ratones , Oxidorreductasas/deficiencia , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Insulina/metabolismo , Transducción de Señal/fisiología
15.
Mol Endocrinol ; 24(11): 2179-92, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20843941

RESUMEN

Insulin receptor substrate-1 (IRS-1) plays a pivotal role in insulin signaling, therefore its degradation is exquisitely regulated. Here, we show that insulin-stimulated degradation of IRS-1 requires the presence of a highly conserved Ser/Thr-rich domain that we named domain involved in degradation of IRS-1 (DIDI). DIDI (amino acids 386-430 of IRS-1) was identified by comparing the intracellular degradation rate of several truncated forms of IRS-1 transfected into CHO cells. The isolated DIDI domain underwent insulin-stimulated Ser/Thr phosphorylation, suggesting that it serves as a target for IRS-1 kinases. The effects of deletion of DIDI were studied in Fao rat hepatoma and in CHO cells expressing Myc-IRS-1(WT) or Myc-IRS-1(Δ386-430). Deletion of DIDI maintained the ability of IRS-1(Δ386-434) to undergo ubiquitination while rendering it insensitive to insulin-induced proteasomal degradation, which affected IRS-1(WT) (80% at 8 h). Consequently, IRS-1(Δ386-434) mediated insulin signaling (activation of Akt and glycogen synthesis) better than IRS-1(WT). IRS-1(Δ386-434) exhibited a significant greater preference for nuclear localization, compared with IRS-1(WT). Higher nuclear localization was also observed when cells expressing IRS-1(WT) were incubated with the proteasome inhibitor MG-132. The sequence of DIDI is conserved more than 93% across species, from fish to mammals, as opposed to approximately 40% homology of the entire IRS-1. These findings implicate DIDI as a novel, highly conserved domain of IRS-1, which mediates its cellular localization, rate of degradation, and biological activity, with a direct impact on insulin signal transduction.


Asunto(s)
Proteínas Sustrato del Receptor de Insulina/química , Proteínas Sustrato del Receptor de Insulina/metabolismo , Insulina/farmacología , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Células CHO , Cricetinae , Cricetulus , Citoprotección/efectos de los fármacos , Ratones , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Fosforilación/efectos de los fármacos , Fosfoserina/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Ratas , Eliminación de Secuencia , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Ubiquitinación/efectos de los fármacos
16.
Diabetes ; 59(9): 2188-97, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20547979

RESUMEN

OBJECTIVE: Cellular stress and proinflammatory cytokines induce phosphorylation of insulin receptor substrate (IRS) proteins at Ser sites that inhibit insulin and IGF-1 signaling. Here, we examined the role of Ser phosphorylation of IRS-2 in mediating the inhibitory effects of proinflammatory cytokines and cellular stress on beta-cell function. RESEARCH DESIGN AND METHODS: Five potential inhibitory Ser sites located proximally to the P-Tyr binding domain of IRS-2 were mutated to Ala. These IRS-2 mutants, denoted IRS-2(5A), and their wild-type controls (IRS-2(WT)) were introduced into adenoviral constructs that were infected into Min6 cells or into cultured murine islets. RESULTS: When expressed in cultured mouse islets, IRS-2(5A) was better than IRS-2(WT) in protecting beta-cells from apoptosis induced by a combination of IL-1beta, IFN-gamma, TNF-alpha, and Fas ligand. Cytokine-treated islets expressing IRS2(5A) secreted significantly more insulin in response to glucose than did islets expressing IRS-2(WT). This could be attributed to the higher transcription of Pdx1 in cytokine-treated islets that expressed IRS-2(5A). Accordingly, transplantation of 200 islets expressing IRS2(5A) into STZ-induced diabetic mice restored their ability to respond to a glucose load similar to naïve mice. In contrast, mice transplanted with islets expressing IRS2(WT) maintained sustained hyperglycemia 3 days after transplantation. CONCLUSIONS: Elimination of a physiological negative feedback control mechanism along the insulin-signaling pathway that involves Ser/Thr phosphorylation of IRS-2 affords protection against the adverse effects of proinflammatory cytokines and improves beta-cell function under stress. Genetic approaches that promote IRS2(5A) expression in pancreatic beta-cells, therefore, could be considered a rational treatment against beta-cell failure after islet transplantation.


Asunto(s)
Proteínas Sustrato del Receptor de Insulina/fisiología , Células Secretoras de Insulina/fisiología , Insulina/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Glucemia/metabolismo , Células CHO , Caspasas/metabolismo , Cricetinae , Cricetulus , Citocinas/farmacología , Diabetes Mellitus Experimental/cirugía , Glucosa/farmacología , Prueba de Tolerancia a la Glucosa , Proteínas de Homeodominio/genética , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/genética , Secreción de Insulina , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/efectos de los fármacos , Células Secretoras de Insulina/metabolismo , Trasplante de Islotes Pancreáticos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Serina/fisiología , Transducción de Señal , Transactivadores/genética , Transfección
17.
J Biol Chem ; 282(25): 18018-18027, 2007 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-17446166

RESUMEN

The Insulin Receptor Substrate (IRS) proteins are key players in insulin signal transduction and are the best studied targets of the insulin receptor. Ser/Thr phosphorylation of IRS proteins negatively modulates insulin signaling; therefore, the identification of IRS kinases and their target Ser phosphorylation sites is of physiological importance. Here we show that in Fao rat hepatoma cells, the IkappaB kinase beta (IKKbeta) is an IRS-1 kinase activated by selected inducers of insulin resistance, including sphingomyelinase, ceramide, and free fatty acids. Moreover, IKKbeta shares a repertoire of seven potential target sites on IRS-1 with protein kinase C zeta (PKCzeta), an IRS-1 kinase activated both by insulin and by inducers of insulin resistance. We further show that mutation of these seven sites (Ser-265, Ser-302, Ser-325, Ser-336, Ser-358, Ser-407, and Ser-408) confers protection from the action of IKKbeta and PKCzeta when they are overexpressed in Fao cells or primary hepatocytes. This enables the mutated IRS proteins to better propagate insulin signaling. These findings suggest that insulin-stimulated IRS kinases such as PKCzeta overlap with IRS kinases triggered by inducers of insulin resistance, such as IKKbeta, to phosphorylate IRS-1 on common Ser sites.


Asunto(s)
Quinasa I-kappa B/metabolismo , Resistencia a la Insulina , Insulina/metabolismo , Fosfoproteínas/fisiología , Proteína Quinasa C/metabolismo , Serina/química , Animales , Ácidos Grasos no Esterificados/metabolismo , Hepatocitos/metabolismo , Humanos , Proteínas Sustrato del Receptor de Insulina , Masculino , Fosfoproteínas/química , Fosforilación , Ratas , Ratas Wistar , Esfingomielina Fosfodiesterasa/metabolismo
18.
Mol Cell Biol ; 24(21): 9668-81, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15485932

RESUMEN

Ser/Thr phosphorylation of insulin receptor substrate (IRS) proteins negatively modulates insulin signaling. Therefore, the identification of serine sites whose phosphorylation inhibit IRS protein functions is of physiological importance. Here we mutated seven Ser sites located proximal to the phosphotyrosine binding domain of insulin receptor substrate 1 (IRS-1) (S265, S302, S325, S336, S358, S407, and S408) into Ala. When overexpressed in rat hepatoma Fao or CHO cells, the mutated IRS-1 protein in which the seven Ser sites were mutated to Ala (IRS-1(7A)), unlike wild-type IRS-1 (IRS-1(WT)), maintained its Tyr-phosphorylated active conformation after prolonged insulin treatment or when the cells were challenged with inducers of insulin resistance prior to acute insulin treatment. This was due to the ability of IRS-1(7A) to remain complexed with the insulin receptor (IR), unlike IRS-1(WT), which underwent Ser phosphorylation, resulting in its dissociation from IR. Studies of truncated forms of IRS-1 revealed that the region between amino acids 365 to 430 is a main insulin-stimulated Ser phosphorylation domain. Indeed, IRS-1 mutated only at S408, which undergoes phosphorylation in vivo, partially maintained the properties of IRS-1(7A) and conferred protection against selected inducers of insulin resistance. These findings suggest that S408 and additional Ser sites among the seven mutated Ser sites are targets for IRS-1 kinases that play a key negative regulatory role in IRS-1 function and insulin action. These sites presumably serve as points of convergence, where physiological feedback control mechanisms, which are triggered by insulin-stimulated IRS kinases, overlap with IRS kinases triggered by inducers of insulin resistance to terminate insulin signaling.


Asunto(s)
Resistencia a la Insulina , Insulina/farmacología , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Fosfotirosina/metabolismo , Serina/metabolismo , Adenoviridae/genética , Animales , Sitios de Unión , Línea Celular , Cricetinae , Activación Enzimática , Regulación Viral de la Expresión Génica , Genes myc/genética , Humanos , Proteínas Sustrato del Receptor de Insulina , Ratones , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutación/genética , Fosfoproteínas/genética , Fosforilación , Fosfoserina/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Serina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA