Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Reprod Dev ; 88(5): 371-375, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33821543

RESUMEN

Intraflagellar transport 27 (IFT27) is a key regulator for spermiogenesis and male fertility in mice. ATP8a1, a protein involved in the translocation of phosphatidylserine and phosphatidylethanolamine across lipid bilayers, is the strongest binding partner of IFT27. To investigate the role of ATP8a1 in spermatogenesis and male fertility, the global Atp8a1 knockout mice were analyzed. All mutant mice were fertile, and sperm count and motility were comparable to the control mice. Examination of testis and epididymis by hematoxylin and eosin staining did not reveal major histologic defects. These observations demonstrate that ATP8a1 is not a major spermatogenesis regulator. Given that a tissue-specific paralogue of ATP8a1, ATP8a2, is present, further studies with double-knockout models are warranted to delineate any compensatory functions of the two proteins.


Asunto(s)
Adenosina Trifosfatasas/fisiología , Fertilidad/fisiología , Proteínas de Transferencia de Fosfolípidos/fisiología , Espermatogénesis/fisiología , Proteínas de Unión al GTP rab/metabolismo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/deficiencia , Adenosina Trifosfatasas/genética , Animales , Epidídimo/ultraestructura , Infertilidad Masculina/genética , Masculino , Lípidos de la Membrana/metabolismo , Ratones , Ratones Noqueados , Fosfatidiletanolaminas/metabolismo , Fosfatidilserinas/metabolismo , Proteínas de Transferencia de Fosfolípidos/química , Proteínas de Transferencia de Fosfolípidos/deficiencia , Proteínas de Transferencia de Fosfolípidos/genética , Dominios Proteicos , Testículo/ultraestructura
2.
Arterioscler Thromb Vasc Biol ; 40(3): 611-623, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31941380

RESUMEN

OBJECTIVE: We tested the hypothesis that enlarged, dysfunctional HDL (high-density lipoprotein) particles contribute to the augmented atherosclerosis susceptibility associated with SR-BI (scavenger receptor BI) deficiency in mice. Approach and Results: We eliminated the ability of HDL particles to fully mature by targeting PLTP (phospholipid transfer protein) functionality. Particle size of the HDL population was almost fully normalized in male and female SR-BI×PLTP double knockout mice. In contrast, the plasma unesterified cholesterol to cholesteryl ester ratio remained elevated. The PLTP deficiency-induced reduction in HDL size in SR-BI knockout mice resulted in a normalized aortic tissue oxidative stress status on Western-type diet. Atherosclerosis susceptibility was-however-only partially reversed in double knockout mice, which can likely be attributed to the fact that they developed a metabolic syndrome-like phenotype characterized by obesity, hypertriglyceridemia, and a reduced glucose tolerance. Mechanistic studies in chow diet-fed mice revealed that the diminished glucose tolerance was probably secondary to the exaggerated postprandial triglyceride response. The absence of PLTP did not affect LPL (lipoprotein lipase)-mediated triglyceride lipolysis but rather modified the ability of VLDL (very low-density lipoprotein)/chylomicron remnants to be cleared from the circulation by the liver through receptors other than SR-BI. As a result, livers of double knockout mice only cleared 26% of the fractional dose of [14C]cholesteryl oleate after intravenous VLDL-like particle injection. CONCLUSIONS: We have shown that disruption of PLTP-mediated HDL maturation reduces SR-BI deficiency-driven atherosclerosis susceptibility in mice despite the induction of proatherogenic metabolic complications in the double knockout mice.


Asunto(s)
Aterosclerosis/prevención & control , HDL-Colesterol/sangre , Metabolismo Energético , Hígado/metabolismo , Síndrome Metabólico/sangre , Proteínas de Transferencia de Fosfolípidos/deficiencia , Receptores Depuradores de Clase B/deficiencia , Animales , Aorta/metabolismo , Aorta/patología , Aterosclerosis/sangre , Aterosclerosis/genética , Aterosclerosis/patología , Ésteres del Colesterol/administración & dosificación , Ésteres del Colesterol/sangre , Modelos Animales de Enfermedad , Femenino , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/genética , Hipertrigliceridemia/sangre , Hipertrigliceridemia/genética , Masculino , Síndrome Metabólico/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/sangre , Obesidad/genética , Proteínas de Transferencia de Fosfolípidos/genética , Placa Aterosclerótica , Receptores Depuradores de Clase B/genética
3.
J Biol Chem ; 293(9): 3386-3398, 2018 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-29348168

RESUMEN

Secretory phospholipases A2 (sPLA2s) are potent components of mammalian innate-immunity antibacterial mechanisms. sPLA2 enzymes attack bacteria by hydrolyzing bacterial membrane phospholipids, causing membrane disorganization and cell lysis. However, most Gram-negative bacteria are naturally resistant to sPLA2 Here we report a novel resistance mechanism to mammalian sPLA2 in Escherichia coli, mediated by a phospholipid repair system consisting of the lysophospholipid transporter LplT and the acyltransferase Aas in the cytoplasmic membrane. Mutation of the lplT or aas gene abolished bacterial lysophospholipid acylation activity and drastically increased bacterial susceptibility to the combined actions of inflammatory fluid components and sPLA2, resulting in bulk phospholipid degradation and loss of colony-forming ability. sPLA2-mediated hydrolysis of the three major bacterial phospholipids exhibited distinctive kinetics and deacylation of cardiolipin to its monoacyl-derivative closely paralleled bacterial death. Characterization of the membrane envelope in lplT- or aas-knockout mutant bacteria revealed reduced membrane packing and disruption of lipid asymmetry with more phosphatidylethanolamine present in the outer leaflet of the outer membrane. Moreover, modest accumulation of lysophospholipids in these mutant bacteria destabilized the inner membrane and rendered outer membrane-depleted spheroplasts much more sensitive to sPLA2 These findings indicated that LplT/Aas inactivation perturbs both the outer and inner membranes by bypassing bacterial membrane maintenance mechanisms to trigger specific interfacial activation of sPLA2 We conclude that the LplT/Aas system is important for maintaining the integrity of the membrane envelope in Gram-negative bacteria. Our insights may help inform new therapeutic strategies to enhance host sPLA2 antimicrobial activity.


Asunto(s)
Aciltransferasas/metabolismo , Membrana Celular/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/citología , Escherichia coli/fisiología , Interacciones Huésped-Patógeno , Fosfolipasas A2/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Fosfolípidos/metabolismo , Aciltransferasas/deficiencia , Animales , Activación Enzimática , Escherichia coli/enzimología , Proteínas de Transferencia de Fosfolípidos/deficiencia
4.
J Lipid Res ; 58(10): 1950-1961, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28765208

RESUMEN

Transmissible spongiform encephalopathies are fatal neurodegenerative diseases with an urgent need for therapeutic and prophylactic strategies. At the time when the blood-mediated transmission of prions was demonstrated, in vitro studies indicated a high binding affinity of the scrapie prion protein (PrPSc) with apoB-containing lipoproteins, i.e., the main carriers of cholesterol in human blood. The aim of the present study was to explore the relationship between circulating cholesterol-containing lipoproteins and the pathogenicity of prions in vivo. We showed that, in mice with a genetically engineered deficiency for the plasma lipid transporter, phospholipid transfer protein (PLTP), abnormally low circulating cholesterol concentrations were associated with a significant prolongation of survival time after intraperitoneal inoculation of the 22L prion strain. Moreover, when circulating cholesterol levels rose after feeding PLTP-deficient mice a lipid-enriched diet, a significant reduction in survival time of mice together with a marked increase in the accumulation rate of PrPSc deposits in their brain were observed. Our results suggest that the circulating cholesterol level is a determinant of prion propagation in vivo and that cholesterol-lowering strategies might be a successful therapeutic approach for patients suffering from prion diseases.


Asunto(s)
Colesterol/sangre , Priones/farmacología , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Femenino , Técnicas de Inactivación de Genes , Ratones , Ratones Endogámicos C57BL , Proteínas de Transferencia de Fosfolípidos/deficiencia , Proteínas de Transferencia de Fosfolípidos/genética , Análisis de Supervivencia
5.
Biochim Biophys Acta ; 1862(9): 1755-65, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27287255

RESUMEN

Type IV ATPases are putative aminophospholipid translocases (APLTs), more commonly known as flippases. A pronounced induction of the flippase Atp8a1 was observed in post-mortem tissue homogenates from the hippocampus and temporal lobe of juvenile autistic subjects compared to age-matched controls. In order to simulate the human data, C57BL/6 mice were allowed to develop after intra-hippocampal injection of recombinant lentivirus expressing Atp8a1 at the early developmental stage of postnatal day 6 (P6). Transmission electron microscopy (TEM) analysis of the lentivirus-Atp8a1 treated (Atp8a1+) mice in adulthood revealed fewer and weaker excitatory synapses in the hippocampal CA1 region compared to mice injected with empty virus. Significant inhibition of the Schaffer collateral pathway was observed in the Atp8a1+ mice in paired-pulse recording (PPR) at 20-ms inter-stimulus interval. In the three-chambered sociability test, the Atp8a1+ mice displayed no preference for an encaged stranger mouse over a novel object, which is a characteristic autistic-like behavior. In sharp contrast, Atp8a1 (-/-) mice displayed a preference for a stranger mouse over the novel object, which is characteristic of neurotypical mouse behavior. However, similar to the Atp8a1+ mice, the Atp8a1 (-/-) mice harbored fewer and weaker excitatory synapses in CA1 compared to wild-type controls, and displayed inhibition at 20-ms inter-stimulus interval in PPR. These findings suggest that both elevated and diminished levels of Atp8a1 during early development are detrimental to brain connectivity, but only elevated Atp8a1 is associated with aberrant social behavior. Mice with augmented levels of Atp8a1 may therefore serve as a potential model in autism research.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Trastorno Autístico/metabolismo , Trastorno Autístico/psicología , Hipocampo/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Adenosina Trifosfatasas/deficiencia , Adenosina Trifosfatasas/genética , Animales , Trastorno Autístico/genética , Conducta Animal , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/ultraestructura , Estudios de Casos y Controles , Niño , Preescolar , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Proteínas de Transferencia de Fosfolípidos/deficiencia , Proteínas de Transferencia de Fosfolípidos/genética , Conducta Social , Sinapsis/metabolismo , Sinapsis/ultraestructura , Lóbulo Temporal/metabolismo
6.
Hum Mol Genet ; 24(19): 5388-403, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26160914

RESUMEN

Phospholipid transfer protein (PLTP) is a widely expressed lipid transfer protein participating in the transport of cholesterol and other lipids in the plasma and peripheral tissues. Recently, elevated amyloid ß (Aß) in young and aged PLTP-deficient brains had been reported. However, the role of PLTP in amyloid precursor protein (APP) processing and Alzheimer's disease (AD) pathology remains elusive. Here we first found that deficiency of PLTP accelerated memory dysfunction in APP/PS1ΔE9 AD model mice at the age of 3 months. Further characterization showed that PLTP deficiency increased soluble Aß peptides, and intracellular accumulation of Aß was illustrated, which might be due to disrupted APP turnover and the enhanced amyloidogenic pathway. Besides, reduced brain-derived neurotrophic factor (BDNF) was found in PLTP-deficient APP/PS1ΔE9 mice, and the BDNF level was negatively correlated with Aß42 content, instead of Aß40 content. In addition, autophagic dysfunction was found in the PLTP-deficient APP/PS1ΔE9 mice. Our data presented a novel model to link phospholipid metabolism to APP processing and also suggested that PLTP played an important role in Aß metabolism and would be useful to further elucidate functions of PLTP in AD susceptibility.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/metabolismo , Trastornos de la Memoria/genética , Proteínas de Transferencia de Fosfolípidos/deficiencia , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Animales , Autofagia , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Humanos , Ratones
7.
Arterioscler Thromb Vasc Biol ; 36(11): 2152-2157, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27634832

RESUMEN

OBJECTIVE: It is known that both platelets and coagulation strongly influence infarct progression after ischemic stroke, but the mechanisms and their interplay are unknown. Our aim was to assess the contribution of the procoagulant platelet surface, and thus platelet-driven thrombin generation, to the progression of thromboinflammation in the ischemic brain. APPROACH AND RESULTS: We present the characterization of a novel platelet and megakaryocyte-specific TMEM16F (anoctamin 6) knockout mouse. Reflecting Scott syndrome, platelets from the knockout mouse had a significant reduction in procoagulant characteristics that altered thrombin and fibrin generation kinetics. In addition, knockout mice showed significant defects in hemostasis and arterial thrombus formation. However, infarct volumes in a model of ischemic stroke were comparable with wild-type mice. CONCLUSIONS: Platelet TMEM16F activity contributes significantly to hemostasis and thrombosis but not cerebral thromboinflammation. These results highlight another key difference between the roles of platelets and coagulation in these processes.


Asunto(s)
Plaquetas/metabolismo , Enfermedades de las Arterias Carótidas/sangre , Encefalitis/sangre , Encefalitis/genética , Hemostasis , Infarto de la Arteria Cerebral Media/sangre , Fosfatidilserinas/sangre , Proteínas de Transferencia de Fosfolípidos/sangre , Trombina/metabolismo , Trombosis/sangre , Animales , Anoctaminas , Coagulación Sanguínea , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/patología , Modelos Animales de Enfermedad , Encefalitis/patología , Fibrina/metabolismo , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/patología , Cinética , Megacariocitos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Transferencia de Fosfolípidos/deficiencia , Proteínas de Transferencia de Fosfolípidos/genética , Activación Plaquetaria , Transducción de Señal , Trombosis/genética , Trombosis/patología
8.
Circulation ; 132(15): 1414-24, 2015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26330411

RESUMEN

BACKGROUND: Platelets are central to the process of hemostasis, rapidly aggregating at sites of blood vessel injury and acting as coagulation nidus sites. On interaction with the subendothelial matrix, platelets are transformed into balloonlike structures as part of the hemostatic response. It remains unclear, however, how and why platelets generate these structures. We set out to determine the physiological relevance and cellular and molecular mechanisms underlying platelet membrane ballooning. METHODS AND RESULTS: Using 4-dimensional live-cell imaging and electron microscopy, we show that human platelets adherent to collagen are transformed into phosphatidylserine-exposing balloonlike structures with expansive macro/microvesiculate contact surfaces, by a process that we termed procoagulant spreading. We reveal that ballooning is mechanistically and structurally distinct from membrane blebbing and involves disruption to the platelet microtubule cytoskeleton and inflation through fluid entry. Unlike blebbing, procoagulant ballooning is irreversible and a consequence of Na(+), Cl(-), and water entry. Furthermore, membrane ballooning correlated with microparticle generation. Inhibition of Na(+), Cl(-), or water entry impaired ballooning, procoagulant spreading, and microparticle generation, and it also diminished local thrombin generation. Human Scott syndrome platelets, which lack expression of Ano-6, also showed a marked reduction in membrane ballooning, consistent with a role for chloride entry in the process. Finally, the blockade of water entry by acetazolamide attenuated ballooning in vitro and markedly suppressed thrombus formation in vivo in a mouse model of thrombosis. CONCLUSIONS: Ballooning and procoagulant spreading of platelets are driven by fluid entry into the cells, and are important for the amplification of localized coagulation in thrombosis.


Asunto(s)
Plaquetas/ultraestructura , Acetazolamida/farmacología , Actomiosina/metabolismo , Amidas/farmacología , Animales , Anoctaminas , Trastornos de la Coagulación Sanguínea/sangre , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Trombosis de las Arterias Carótidas/sangre , Trombosis de las Arterias Carótidas/inducido químicamente , Trombosis de las Arterias Carótidas/tratamiento farmacológico , Adhesión Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Forma de la Célula/efectos de los fármacos , Forma de la Célula/fisiología , Tamaño de la Célula/efectos de los fármacos , Micropartículas Derivadas de Células , Cloruros/metabolismo , Colágeno , Citocalasina D/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Ratones , Microtúbulos/efectos de los fármacos , Proteínas de Transferencia de Fosfolípidos/deficiencia , Proteínas de Transferencia de Fosfolípidos/fisiología , Piridinas/farmacología , Sodio/metabolismo , Trombina/biosíntesis , Trombosis/prevención & control , Agua/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 35(2): 316-22, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25477345

RESUMEN

OBJECTIVE: Phospholipid transfer protein (PLTP) is highly expressed in adipose tissues. Thus, the effect of adipose tissue PLTP on plasma lipoprotein metabolism was examined. APPROACH AND RESULTS: We crossed PLTP-Flox-ΔNeo and adipocyte protein 2 (aP2)-Cre recombinase (Cre) transgenic mice to create PLTP-Flox-ΔNeo/aP2-Cre mice that have a 90 and a 60% reduction in PLTP mRNA in adipose tissue and macrophages, respectively. PLTP ablation resulted in a significant reduction in plasma PLTP activity (22%), high-density lipoprotein-cholesterol (21%), high-density lipoprotein-phospholipid (20%), and apolipoprotein A-I (33%) levels, but had no effect on nonhigh-density lipoprotein levels in comparison with those of PLTP-Flox-ΔNeo controls. To eliminate possible effects of PLTP ablation by macrophages, we lethally irradiated PLTP-Flox-ΔNeo/aP2-Cre mice and PLTP-Flox-ΔNeo mice, and then transplanted wild-type mouse bone marrow into them to create wild-type→PLTP-Flox-ΔNeo/aP2-Cre and wild-type→PLTP-Flox-ΔNeo mice. Thus, we constructed a mouse model (wild-type→PLTP-Flox-ΔNeo/aP2-Cre) with PLTP deficiency in adipocytes but not in macrophages. These knockout mice also showed significant decreases in plasma PLTP activity (19%) and cholesterol (18%), phospholipid (17%), and apolipoprotein A-I (26%) levels. To further investigate the mechanisms behind the reduction in plasma apolipoprotein A-I and high-density lipoprotein lipids, we measured apolipoprotein A-I-mediated cholesterol efflux in adipose tissue explants and found that endogenous and exogenous PLTP significantly increased cholesterol efflux from the explants. CONCLUSIONS: Adipocyte PLTP plays a small but significant role in plasma PLTP activity and promotes cholesterol efflux from adipose tissues.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Lipoproteínas HDL/sangre , Proteínas de Transferencia de Fosfolípidos/metabolismo , Tejido Adiposo/citología , Animales , Apolipoproteína A-I/sangre , Trasplante de Médula Ósea , Células Cultivadas , Colesterol/sangre , Proteínas de Unión a Ácidos Grasos/genética , Genotipo , Integrasas/genética , Macrófagos/metabolismo , Ratones Noqueados , Fenotipo , Proteínas de Transferencia de Fosfolípidos/deficiencia , Proteínas de Transferencia de Fosfolípidos/genética , Fosfolípidos/sangre , Factores de Tiempo , Técnicas de Cultivo de Tejidos
10.
Cell Physiol Biochem ; 37(5): 1934-44, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26584292

RESUMEN

BACKGROUND/AIMS: The ubiquitously expressed Ca2+ Activated Cl- Channel Ano6 participates in the stimulation of cell membrane scrambling. Defective Ano6 underlies the Scott syndrome, an inherited bleeding disorder with impaired scrambling of plasma membrane phospholipids. At least in theory, the bleeding disorder of Scott syndrome may result from impaired platelet function. Activators of platelets include thrombin and collagen related peptide (CRP), which trigger increase of cytosolic Ca2+-activity ([Ca2+]i), production of reactive oxygen species (ROS), degranulation, integrin activation, as well as cell shrinkage and phospholipid scrambling of the cell membrane. The present study thus explored whether Ano6 modifies activation-induced alterations of cytosolic Ca2+-activity ([Ca2+]i), degranulation (P-selectin exposure), integrin activation, phosphatidylserine exposure on the platelet surface and platelet volume. METHODS: Platelets from mice lacking Ano6 (ano6-/-) were compared to platelets from corresponding wild-type mice (ano6+/+). [Ca2+]i was estimated from Fluo-3 fluorescence, ROS from DCFDA fluorescence, degranulation from P-selectin abundance, integrin activation from αIIbß3-integrin abundance, phosphatidylserine abundance from annexin-V-binding, and cell volume from forward scatter. RESULTS: Platelet number in blood was slightly higher in ano6-/- mice than in ano6+/+ mice. Without activation [Ca2+]i and volume were similar in ano6-/- and ano6+/+ platelets as well as ROS abundance, P-selectin abundance, αIIbß3 integrin activation, and phosphatidylserine exposure were negligible in both genotypes. Thrombin (0.01 U/ml) and CRP (2 or 5 µg/ml) increased [Ca2+]i, ROS abundance, platelet degranulation, αIIbß3 integrin activation, and triggered annexin-V-binding as well as cell shrinkage, all effects less pronounced in ano6-/- than in ano6+/+ platelets. CONCLUSIONS: Genetic knockout of Ano6 blunts thrombin- and CRP-induced activation and apoptosis of blood platelets.


Asunto(s)
Apoptosis , Plaquetas/metabolismo , Activación Plaquetaria , Compuestos de Anilina/química , Animales , Anoctaminas , Apoptosis/efectos de los fármacos , Plaquetas/citología , Proteína C-Reactiva/farmacología , Calcio/análisis , Ratones , Ratones Noqueados , Selectina-P/metabolismo , Fosfatidilserinas/metabolismo , Proteínas de Transferencia de Fosfolípidos/deficiencia , Proteínas de Transferencia de Fosfolípidos/genética , Activación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Especies Reactivas de Oxígeno/análisis , Especies Reactivas de Oxígeno/metabolismo , Trombina/farmacología , Xantenos/química
11.
Arterioscler Thromb Vasc Biol ; 34(12): 2537-44, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25324570

RESUMEN

OBJECTIVE: Phospholipid transfer protein (PLTP) accelerates the development of atherosclerosis in mouse models. We examined the role of PLTP in atherosclerotic plaque stability. APPROACH AND RESULTS: We prepared apolipoprotein E and PLTP double-knockout (PLTP(-/-)ApoE(-/-)) mice. PLTP deficiency significantly decreased lesion size and reduced monocyte/macrophage infiltration, as well as macrophage apoptosis in lesion areas. Moreover, it increased fibrous content in plaques, which suggests that PLTP may affect atherosclerotic plaque stability. Importantly, PLTP overexpression mediated by adenovirus had the reverse effect. It promoted the accumulation of reactive oxygen species in macrophages, which could lead to cell apoptosis and increased the production of inflammatory cytokines and chemokines. PLTP overexpression could promote receptor-interacting protein 3 recruitment of macrophages in cytoplasm, which could induce reactive oxygen species, thus inducing atherogenesis. CONCLUSIONS: PLTP plays an important role in modulating the stability of atherosclerotic plaques. The receptor-interacting protein 3- reactive oxygen species signal pathway could be involved in this PLTP-mediated process.


Asunto(s)
Proteínas de Transferencia de Fosfolípidos/metabolismo , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patología , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Apoptosis , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Estenosis Carotídea/genética , Estenosis Carotídea/metabolismo , Estenosis Carotídea/patología , Caspasa 3/metabolismo , Quimiocinas/sangre , Citocinas/sangre , Lípidos/sangre , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas de Transferencia de Fosfolípidos/deficiencia , Proteínas de Transferencia de Fosfolípidos/genética , Placa Aterosclerótica/genética , Especies Reactivas de Oxígeno/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal , Regulación hacia Arriba
12.
Dig Dis ; 33(3): 314-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26045263

RESUMEN

P4 ATPases are lipid flippases and transport phospholipids from the exoplasmic to the cytosolic leaflet of biological membranes. Lipid flipping is important for the biogenesis of transport vesicles. Recently it was shown that loss of the P4 ATPases ATP8B1 and ATP11C are associated with severe Cholestatic liver disease. Mutation of ATP8B1 cause progressive familial Intrahepatic Cholestasis type 1 (PFIC1)and benign recurrent intrahepatic cholestasis type 1 (BRIC 1). From our observations we hypothesized that ATP8B1 deficiency causes a phospholipids randomization at the canalicular membrane, which results in extraction of cholesterol due to increase sensitivity of the canalicular membrane. Deficiency of ATP11C causes conjugated hyperbilirubinemia. In our preliminary result we observed accumulation of unconjugated bile salts in Atp11c deficient mice probably because of regulation in the expression or function of OATP1B2. Similar to ATP8B1, ATP11C have regulation on membrane transporters.


Asunto(s)
Adenosina Trifosfatasas/genética , Colestasis Intrahepática/genética , Proteínas de Transferencia de Fosfolípidos/genética , Adenosina Trifosfatasas/deficiencia , Animales , Ácidos Cólicos/sangre , Ácidos Cólicos/genética , Hepatocitos/fisiología , Humanos , Hiperbilirrubinemia/genética , Ratones , Mutación , Proteínas de Transferencia de Fosfolípidos/deficiencia , Errores Congénitos del Metabolismo Esteroideo/genética
13.
Am J Pathol ; 183(3): 975-86, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23830874

RESUMEN

Plasma phospholipid transfer protein (PLTP) increases the circulating levels of proatherogenic lipoproteins, accelerates blood coagulation, and modulates inflammation. The role of PLTP in the development of abdominal aortic aneurysm (AAA) was investigated by using either a combination of mechanical and elastase injury at one site of mouse aorta (elastase model) or continuous infusion of angiotensin II in hyperlipidemic ApoE-knockout mice (Ang II model). With the elastase model, complete PLTP deficiency was associated with a significantly lower incidence and a lesser degree of AAA expansion. With the Ang II model, findings were consistent with those in the elastase model, with a lower severity grade in PLTP-deficient mice, an intermediate phenotype in PLTP-deficient heterozygotes, and a blunted effect of the PLTP-deficient trait when restricted to bone marrow-derived immune cells. The protective effect of whole-body PLTP deficiency in AAA was illustrated further by a lesser degree of adventitia expansion, reduced elastin degradation, fewer recruited macrophages, and less smooth muscle cell depletion in PLTP-deficient than in wild-type mice, as evident from comparative microscopic analysis of aorta sections. Finally, cumulative evidence supports the association of PLTP deficiency with reduced expression and activity levels of matrix metalloproteinases, known to degrade elastin and collagen. We conclude that PLTP can play a significant role in the pathophysiology of AAA.


Asunto(s)
Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Proteínas de Transferencia de Fosfolípidos/deficiencia , Proteínas de Transferencia de Fosfolípidos/metabolismo , Angiotensina II , Animales , Aorta/patología , Aneurisma de la Aorta Abdominal/complicaciones , Apolipoproteínas E/deficiencia , Linfocitos T CD4-Positivos/metabolismo , Citocinas/metabolismo , Elastina/metabolismo , Inflamación/complicaciones , Inflamación/patología , Hígado/metabolismo , Hígado/patología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Elastasa Pancreática
14.
Arterioscler Thromb Vasc Biol ; 33(9): 2058-64, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23846500

RESUMEN

OBJECTIVE: The liver is one of the critical organs for lipoprotein metabolism and a major source for phospholipid transfer protein (PLTP) expression. The effect of liver-specific PLTP deficiency on plasma lipoprotein production and metabolism in mice was investigated. APPROACH AND RESULTS: We created a liver-specific PLTP-deficient mouse model. We measured plasma high-density lipoprotein (HDL) and apolipoprotein B (apoB)-containing lipoprotein (or non-HDL) levels and their production rates. We found that hepatic ablation of PLTP leads to a significant decrease in plasma PLTP activity, HDL lipids, non-HDL lipids, apoAI, and apoB levels. In addition, nuclear magnetic resonance examination of lipoproteins showed that the deficiency decreases HDL and apoB-containing lipoprotein particle numbers, as well as very low-density lipoprotein particle size, which was confirmed by electron microscopy. Moreover, HDL particles from the deficient mice are lipid-poor ones. To unravel the mechanism, we evaluated the apoB and triglyceride production rates. We found that hepatic PLTP deficiency significantly decreases apoB and triglyceride secretion rates. To investigate the role of liver PLTP on HDL production, we set up primary hepatocyte culture studies and found that the PLTP-deficient hepatocytes produce less nascent HDL. Furthermore, we found that exogenous PLTP promotes nascent HDL production through an ATP-binding cassette A 1-mediated pathway. CONCLUSIONS: Liver-specific PLTP deficiency significantly reduces plasma HDL and apoB-containing lipoprotein levels. Reduction of production rates of both particles is one of the mechanisms.


Asunto(s)
Hepatocitos/metabolismo , Lipoproteínas HDL/metabolismo , Hígado/metabolismo , Proteínas de Transferencia de Fosfolípidos/deficiencia , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Apolipoproteína A-I/sangre , Apolipoproteínas B/sangre , Biomarcadores/sangre , Células Cultivadas , Regulación hacia Abajo , Lipoproteínas VLDL/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica , Proteínas de Transferencia de Fosfolípidos/genética , Cultivo Primario de Células , Triglicéridos/sangre
15.
Int J Biol Macromol ; 267(Pt 2): 131240, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38583827

RESUMEN

Lipids are intimately related to the unique flavor and nutritional values of goat milk. MicroRNAs (miRNA) participate in the regulation of various biological functions, including the synthesis and degradation of lipids. Several studies have shown that miR-103 is involved in the regulation of lipid metabolism, however, the molecular mechanism by which miR-103 regulates lipid metabolism in goat mammary gland is poorly understood. In this study, miR-103 was knocked out in goat mammary epithelial cells (GMECs) by CRISPR/Cas9, and the accumulation of lipid droplets, triglycerides, and cholesterol in the cells was suppressed subsequently. Overexpression or knockdown of miR-103-5p and miR-103-3p in GMECs revealed that it was miR-103-5p that promoted lipid accumulation but not miR-103-3p. In addition, Pantothenate Kinase 3 (PANK3), the host gene of miR-103, and Phospholipid Scramblase 4 (PLSCR4) were identified as the target genes of miR-103-5p by dual fluorescein and miRNA pulldown. Furthermore, we identified that cellular lipid levels were negatively regulated by PANK3 and PLSCR4. Lastly, in miR-103 knockout GMECs, the knockdown of PANK and PLSCR4 rescued the lipid accumulation. These findings suggest that miR-103-5p promotes lipid accumulation by targeting PLSCR4 and the host gene PANK3 in GMECs, providing new insights for the regulation of goat milk lipids via miRNAs.


Asunto(s)
Células Epiteliales , Cabras , Metabolismo de los Lípidos , Glándulas Mamarias Animales , MicroARNs , Fosfotransferasas (Aceptor de Grupo Alcohol) , Animales , MicroARNs/genética , MicroARNs/metabolismo , Cabras/genética , Metabolismo de los Lípidos/genética , Células Epiteliales/metabolismo , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/citología , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Femenino , Proteínas de Transferencia de Fosfolípidos/genética , Proteínas de Transferencia de Fosfolípidos/metabolismo , Proteínas de Transferencia de Fosfolípidos/deficiencia , Regulación hacia Arriba/genética , Gotas Lipídicas/metabolismo , Regulación de la Expresión Génica , Triglicéridos/metabolismo
16.
J Neurochem ; 120(2): 302-13, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22007859

RESUMEN

The molecule responsible for the enzyme activity plasma membrane (PM) aminophospholipid translocase (APLT), which catalyzes phosphatidylserine (PS) translocation from the outer to the inner leaflet of the plasma membrane, is unknown in mammals. A Caenorhabditis elegans study has shown that ablation of transbilayer amphipath transporter-1 (TAT-1), which is an ortholog of a mammalian P-type ATPase, Atp8a1, causes PS externalization in the germ cells. We demonstrate here that the hippocampal cells of the dentate gyrus, and Cornu Ammonis (CA1, CA3) in mice lacking Atp8a1 exhibit a dramatic increase in PS externalization. Although their hippocampi showed no abnormal morphology or heightened apoptosis, these mice displayed increased activity and a marked deficiency in hippocampus-dependent learning, but no hyper-anxiety. Such observations indicate that Atp8a1 plays a crucial role in PM-APLT activity in the neuronal cells. In corroboration, ectopic expression of Atp8a1 but not its close homolog, Atp8a2, caused an increase in the population (V(max) ) of PM-APLT without any change in its signature parameter K(m) in the neuronal N18 cells. Conversely, expression of a P-type phosphorylation-site mutant of Atp8a1 (Atp8a1*) caused a decrease in V(max) of PM-APLT without significantly altering its K(m) . The Atp8a1*-expressing N18 cells also exhibited PS externalization without apoptosis. Together, our data strongly indicate that Atp8a1 plays a central role in the PM-APLT activity of some mammalian cells, such as the neuronal N18 and hippocampal cells.


Asunto(s)
Adenosina Trifosfatasas/deficiencia , Regulación de la Expresión Génica/genética , Hipocampo/metabolismo , Aprendizaje por Laberinto/fisiología , Fosfatidilserinas/metabolismo , Proteínas de Transferencia de Fosfolípidos/deficiencia , Animales , Anexina A5/metabolismo , Membrana Celular/metabolismo , Femenino , Hipocampo/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Técnicas de Cultivo de Órganos , Proteínas de Transferencia de Fosfolípidos/metabolismo , Fosforilación/genética , Transporte de Proteínas/genética
17.
Am J Pathol ; 178(5): 2058-65, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21514421

RESUMEN

In the tear fluid the outermost part facing the tear-air interface is composed of lipids preventing evaporation of the tears. Phospholipid transfer protein (PLTP) mediates phospholipid transfer processes between serum lipoproteins and is also a normal component of human tears. To study whether PLTP plays any functional role in tear fluid we investigated PLTP-deficient mice, applying functional and morphologic analyses under normal housing and experimentally induced dry eye conditions. Aqueous tear fluid production, corneal epithelial morphology, barrier function, and occludin expression were assessed. In mice with a full deficiency of functional PLTP enhanced corneal epithelial damage, increased corneal permeability to carboxyfluorescein, and decreased corneal epithelial occludin expression were shown. These pathologic signs were worsened by experimentally induced dry eye both in wild-type and PLTP knock-out mice. Deficiency in the production of tear PLTP in mice is accompanied by corneal epithelial damage, a feature that is typical in human dry eye syndrome (DES). To complement animal experiments we collected tear fluid from human dry eye patients as well as healthy control subjects. Increased tear fluid PLTP activity was observed among DES patients. In conclusion, the presence of PLTP in tear fluid appears to be essential for maintaining a healthy and functional ocular surface. Increased PLTP activity in human tear fluid in DES patients suggests an ocular surface protective role for this lipid transfer protein.


Asunto(s)
Síndromes de Ojo Seco/metabolismo , Epitelio Corneal/metabolismo , Proteínas de Transferencia de Fosfolípidos/metabolismo , Lágrimas/metabolismo , Adulto , Anciano , Animales , Western Blotting , Permeabilidad de la Membrana Celular/fisiología , Síndromes de Ojo Seco/patología , Epitelio Corneal/patología , Femenino , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Proteínas de Transferencia de Fosfolípidos/deficiencia , Lágrimas/química , Uniones Estrechas/metabolismo
18.
Arterioscler Thromb Vasc Biol ; 30(12): 2452-7, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20864671

RESUMEN

OBJECTIVE: Earlier in vitro studies suggested a putative role for the plasma phospholipid transfer protein (PLTP) in the modulation of blood coagulation. The effect of PLTP expression on blood coagulation under both basal and oxidative stress conditions was compared here in wild-type and PLTP-deficient (PLTP-/-) mice. METHODS AND RESULTS: Under basal conditions, PLTP deficiency was associated with an extended tail bleeding time despite a significant depletion of vascular α-tocopherol content and an impairment of endothelial function. When acute oxidative stress was generated in vivo in the brain vasculature, the steady state levels of oxidized lipid derivatives, the extent of blood vessel occlusion, and the volume of ischemic lesions were more severe in wild-type than in PLTP-/- mice. CONCLUSIONS: In addition to its recognized hyperlipidemic, proinflammatory, and proatherogenic properties, PLTP increases blood coagulation and worsens the extent of ischemic lesions in response to acute oxidative stress. Thus, PLTP arises here as a cardiovascular risk factor for the late thrombotic events occurring in the acute phase of atherosclerosis.


Asunto(s)
Coagulación Sanguínea , Infarto Cerebral/prevención & control , Endotelio Vascular/metabolismo , Trombosis Intracraneal/prevención & control , Estrés Oxidativo , Proteínas de Transferencia de Fosfolípidos/deficiencia , Animales , Tiempo de Sangría , Infarto Cerebral/sangre , Infarto Cerebral/genética , Infarto Cerebral/patología , Infarto Cerebral/fisiopatología , Relación Dosis-Respuesta a Droga , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Trombosis Intracraneal/sangre , Trombosis Intracraneal/genética , Trombosis Intracraneal/patología , Trombosis Intracraneal/fisiopatología , Ácidos Linoleicos/metabolismo , Metabolismo de los Lípidos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Proteínas de Transferencia de Fosfolípidos/genética , Vasodilatadores/farmacología , alfa-Tocoferol/sangre
19.
Nat Cell Biol ; 23(3): 268-277, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33664495

RESUMEN

The sympathetic nervous system-catecholamine-uncoupling protein 1 (UCP1) axis plays an essential role in non-shivering adaptive thermogenesis. However, whether there exists a direct effector that physically connects catecholamine signalling to UCP1 in response to acute cold is unknown. Here we report that outer mitochondrial membrane-located AIDA is phosphorylated at S161 by the catecholamine-activated protein kinase A (PKA). Phosphorylated AIDA translocates to the intermembrane space, where it binds to and activates the uncoupling activity of UCP1 by promoting cysteine oxidation of UCP1. Adipocyte-specific depletion of AIDA abrogates UCP1-dependent thermogenesis, resulting in hypothermia during acute cold exposure. Re-expression of S161A-AIDA, unlike wild-type AIDA, fails to restore the acute cold response in Aida-knockout mice. The PKA-AIDA-UCP1 axis is highly conserved in mammals, including hibernators. Denervation of the sympathetic postganglionic fibres abolishes cold-induced AIDA-dependent thermogenesis. These findings uncover a direct mechanistic link between sympathetic input and UCP1-mediated adaptive thermogenesis.


Asunto(s)
Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/inervación , Proteínas de Transferencia de Fosfolípidos/metabolismo , Sistema Nervioso Simpático/fisiología , Termogénesis , Proteína Desacopladora 1/metabolismo , Adiponectina/genética , Adiponectina/metabolismo , Animales , Células Cultivadas , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Metabolismo Energético , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Proteínas de Transferencia de Fosfolípidos/deficiencia , Proteínas de Transferencia de Fosfolípidos/genética , Fosforilación , Transducción de Señal , Proteína Desacopladora 1/deficiencia , Proteína Desacopladora 1/genética
20.
J Lipid Res ; 51(10): 2993-3002, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20543142

RESUMEN

A high saturated fat diet induces free cholesterol and phospholipid accumulation in the plasma of phospholipid transfer protein (Pltp)-deficient mice. In this study, we examined the atherogenic consequence of this phenomenon and investigated the possible mechanism(s). Pltp KO/Apoe KO mice that were fed a coconut oil-enriched high-fat diet (COD) for 7 weeks had higher plasma free cholesterol (149%), phospholipids (15%), and sphingomyelin (54%) than Apoe KO controls. In contrast to chow-fed animals, COD-fed Pltp KO/Apoe KO mice had the same atherosclerotic lesion size as that of Apoe KO mice. Similar to Pltp KO mice, plasma from COD-fed Pltp KO/Apoe KO mice contained VLDL/LDL-sized lamellar particles. Bile measurement indicated that COD-fed Pltp KO mice have 33% less hepatic cholesterol output than controls. In conclusion, COD-fed, Pltp-deficient mice are no longer protected from atherosclerosis and have impaired biliary lipid secretion, which is associated with free cholesterol and phospholipid accumulation.


Asunto(s)
Aterosclerosis/metabolismo , Colesterol/sangre , Proteínas de Transferencia de Fosfolípidos/genética , Fosfolípidos/sangre , Animales , Aterosclerosis/sangre , Aterosclerosis/etiología , Dieta , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica , Proteínas de Transferencia de Fosfolípidos/deficiencia , Proteínas de Transferencia de Fosfolípidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA