Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Biol Chem ; 299(6): 104812, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37172724

RESUMEN

T-cell acute lymphoblastic leukemia (T-ALL) is one of the deadliest and most aggressive hematological malignancies, but its pathological mechanism in controlling cell survival is not fully understood. Oculocerebrorenal syndrome of Lowe is a rare X-linked recessive disorder characterized by cataracts, intellectual disability, and proteinuria. This disease has been shown to be caused by mutation of oculocerebrorenal syndrome of Lowe 1 (OCRL1; OCRL), encoding a phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] 5-phosphatase involved in regulating membrane trafficking; however, its function in cancer cells is unclear. Here, we uncovered that OCRL1 is overexpressed in T-ALL cells, and knockdown of OCRL1 results in cell death, indicating the essential role of OCRL in controlling T-ALL cell survival. We show OCRL is primarily localized in the Golgi and can translocate to plasma membrane (PM) upon ligand stimulation. We found OCRL interacts with oxysterol-binding protein-related protein 4L, which facilitates OCRL translocation from the Golgi to the PM upon cluster of differentiation 3 stimulation. Thus, OCRL represses the activity of oxysterol-binding protein-related protein 4L to prevent excessive PI(4,5)P2 hydrolysis by phosphoinositide phospholipase C ß3 and uncontrolled Ca2+ release from the endoplasmic reticulum. We propose OCRL1 deletion leads to accumulation of PI(4,5)P2 in the PM, disrupting the normal Ca2+ oscillation pattern in the cytosol and leading to mitochondrial Ca2+ overloading, ultimately causing T-ALL cell mitochondrial dysfunction and cell death. These results highlight a critical role for OCRL in maintaining moderate PI(4,5)P2 availability in T-ALL cells. Our findings also raise the possibility of targeting OCRL1 to treat T-ALL disease.


Asunto(s)
Membrana Celular , Fosfatidilinositol 4,5-Difosfato , Monoéster Fosfórico Hidrolasas , Leucemia-Linfoma Linfoblástico de Células T Precursoras , Linfocitos T , Humanos , Membrana Celular/metabolismo , Supervivencia Celular , Hidrólisis , Síndrome Oculocerebrorrenal/enzimología , Síndrome Oculocerebrorrenal/genética , Fosfatidilinositol 4,5-Difosfato/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Linfocitos T/citología , Linfocitos T/inmunología , Monoéster Fosfórico Hidrolasas/biosíntesis , Monoéster Fosfórico Hidrolasas/deficiencia , Monoéster Fosfórico Hidrolasas/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Aparato de Golgi/metabolismo , Ligandos , Transporte de Proteínas , Señalización del Calcio , Mitocondrias/metabolismo , Mitocondrias/patología , Citosol/metabolismo
2.
Blood ; 139(7): 1052-1065, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-34797912

RESUMEN

Human T-cell leukemia virus 1 (HTLV-1) causes adult T-cell leukemia (ATL), but the mechanism underlying its initiation remains elusive. In this study, ORP4L was expressed in ATL cells but not in normal T-cells. ORP4L ablation completely blocked T-cell leukemogenesis induced by the HTLV-1 oncoprotein Tax in mice, whereas engineering ORP4L expression in T-cells resulted in T-cell leukemia in mice, suggesting the oncogenic properties and prerequisite of ORP4L promote the initiation of T-cell leukemogenesis. For molecular insight, we found that loss of miR-31 caused by HTLV-1 induced ORP4L expression in T-cells. ORP4L interacts with PI3Kδ to promote PI(3,4,5)P3 generation, contributing to AKT hyperactivation; NF-κB-dependent, p53 inactivation-induced pro-oncogene expression; and T-cell leukemogenesis. Consistently, ORP4L ablation eliminates human ATL cells in patient-derived xenograft ATL models. These results reveal a plausible mechanism of T-cell deterioration by HTLV-1 that can be therapeutically targeted.


Asunto(s)
Carcinogénesis/patología , Regulación Leucémica de la Expresión Génica , Infecciones por HTLV-I/complicaciones , Virus Linfotrópico T Tipo 1 Humano/aislamiento & purificación , Leucemia-Linfoma de Células T del Adulto/patología , Receptores de Esteroides/metabolismo , Linfocitos T/inmunología , Animales , Apoptosis , Carcinogénesis/inmunología , Carcinogénesis/metabolismo , Proliferación Celular , Productos del Gen tax , Infecciones por HTLV-I/virología , Humanos , Leucemia-Linfoma de Células T del Adulto/genética , Leucemia-Linfoma de Células T del Adulto/metabolismo , Leucemia-Linfoma de Células T del Adulto/virología , Ratones , Pronóstico , Receptores de Esteroides/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
3.
J Immunol ; 204(5): 1134-1145, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31953353

RESUMEN

Phospholipase C (PLC) isoforms play central roles in signaling cascades by cleaving PIP2 into the second messengers IP3 and DAG. In this study, to our knowledge, we uncover that ORP5L interacts physically with PLCγ1 in T cells, extracts PIP2 from the plasma membrane via its ORD domain (OSBP-related domain), presents it to PLCγ1 (enabling IP3 generation), and eventually maintains intracellular Ca2+ homeostasis. Through this mechanism, ORP5L promotes T cell proliferation in a Ca2+-activated NFAT2-dependent manner. To our knowledge, our study uncovers a new key function of ORP5L as a critical cofactor for PLCγ1 catalysis and its crucial role in human T cell proliferation.


Asunto(s)
Señalización del Calcio/inmunología , Proliferación Celular , Inositol 1,4,5-Trifosfato/inmunología , Fosfatidilinositol 4,5-Difosfato/inmunología , Receptores de Esteroides/inmunología , Femenino , Humanos , Hidrólisis , Masculino , Fosfolipasa C gamma/inmunología
4.
FASEB J ; 34(11): 14671-14694, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32914503

RESUMEN

Oxysterol-binding protein-related protein 2 (ORP2), a cholesterol-PI(4,5)P2 countercurrent transporter, was recently identified as a novel regulator of plasma membrane (PM) cholesterol and PI(4,5)P2 content in HeLa cells. Here, we investigate the role of ORP2 in endothelial cell (EC) cholesterol and PI(4,5)P2 distribution, angiogenic signaling, and angiogenesis. We show that ORP2 knock-down modifies the distribution of cholesterol accessible to a D4H probe, between late endosomes and the PM. Depletion of ORP2 from ECs inhibits their angiogenic tube formation capacity, alters the gene expression of angiogenic signaling pathways such as VEGFR2, Akt, mTOR, eNOS, and Notch, and reduces EC migration, proliferation, and cell viability. We show that ORP2 regulates the integrity of VEGFR2 at the PM in a cholesterol-dependent manner, the depletion of ORP2 resulting in proteolytic cleavage by matrix metalloproteinases, and reduced activity of VEGFR2 and its downstream signaling. We demonstrate that ORP2 depletion increases the PM PI(4,5)P2 coincident with altered F-actin morphology, and reduces both VEGFR2 and cholesterol in buoyant raft membranes. Moreover, ORP2 knock-down suppresses the expression of the lipid raft-associated proteins VE-cadherin and caveolin-1. Analysis of the retinal microvasculature in ORP2 knock-out mice generated during this study demonstrates the subtle alterations of morphology characterized by reduced vessel length and increased density of tip cells and perpendicular sprouts. Gene expression changes in the retina suggest disturbance of sterol homeostasis, downregulation of VE-cadherin, and a putative disturbance of Notch signaling. Our data identifies ORP2 as a novel regulator of EC cholesterol and PI(4,5)P2 homeostasis and cholesterol-dependent angiogenic signaling.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/metabolismo , Neovascularización Fisiológica , Receptores de Esteroides/metabolismo , Transducción de Señal , Actinas/metabolismo , Antígenos CD/metabolismo , Cadherinas/metabolismo , Caveolinas/metabolismo , Membrana Celular/metabolismo , Movimiento Celular , Endosomas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Metaloproteinasas de la Matriz/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosfatidilinositol 4,5-Difosfato/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Notch/metabolismo , Receptores de Esteroides/genética , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
5.
FASEB J ; 33(1): 545-556, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30004795

RESUMEN

The enhanced expression of miR-31 has been observed in many human malignancies including lung cancer, and this microRNA regulates several aspects of oncogenesis. However, the role of miR-31-5p in energy metabolism remains elusive. Here, we confirm that H1299 and A549 cells, 2 lung cancer cell lines, relay on aerobic glycolysis as main source of ATP. Inhibition of miR-31-5p leads to decreased glycolysis and ATP production, while miR-31-5p overexpression increases them. Hypoxia inducible factor 1 (HIF-1) up-regulates the expression of glycolytic enzymes, and the HIF-1α inhibitor (FIH) inhibits HIF-1 activity. Because FIH is a direct target of miR-31-5p, inhibition of miR-31-5p results in enhanced FIH expression and suppression of HIF-1 signaling, while overexpression of miR-31-5p has the opposite effects. Via this mechanism, miR-31-5p up-regulates aerobic glycolytic genes and maintains energy homeostasis. To further validate the mechanism of miR-31-5p in glycolysis regulation, we show that overexpression or knockdown of FIH rescued the effects of miR-31-5p or miR-31-5p inhibitor on HIF activation and its target gene expression, respectively. Finally, by means of an A549 cell xenograft mouse model, we demonstrate that the miR-31-5p promotes cell proliferation via enhancing glycolysis. In summary, this study reveals that miR-31-5p promotes the Warburg effect via direct targeting of FIH.-Zhu, B., Cao, X., Zhang, W., Pan, G., Yi, Q., Zhong, W., Yan, D. MicroRNA-31-5p enhances the Warburg effect via targeting FIH.


Asunto(s)
Glucólisis , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ácido Láctico/metabolismo , Neoplasias Pulmonares/patología , MicroARNs/genética , Oxigenasas de Función Mixta/metabolismo , Ácido Pirúvico/metabolismo , Proteínas Represoras/metabolismo , Animales , Apoptosis , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Oxigenasas de Función Mixta/genética , Proteínas Represoras/genética , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
FASEB J ; 33(12): 13852-13865, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31648575

RESUMEN

Oxysterol-binding protein-related protein (ORP) 4L acts as a scaffold protein assembling CD3-ε, G-αq/11, and PLC-ß3 into a complex at the plasma membrane that mediates inositol (1,4,5)-trisphosphate (IP3)-induced endoplasmic reticulum (ER) Ca2+ release and oxidative phosphorylation in T-cell acute lymphoblastic leukemia cells. Here, we offer new evidence that ORP4L interacts with the carboxyl terminus of the IP3 receptor type 1 (ITPR1) in Jurkat T cells. ORP4L enables IP3 binding to ITPR1; a truncated construct that lacks the ITPR1-binding region retains the ability to increase IP3 production but fails to mediate IP3 and ITPR1 binding. In association with this ability of ORP4L, it enhances Ca2+ release from the ER and subsequent cytosolic and mitochondrial parallel Ca2+ spike oscillations that stimulate mitochondrial energetics and thus maintains cell survival. These data support a novel model in which ORP4L is a cofactor of ITPR1, which increases ITPR1 sensitivity to IP3 and enables ER Ca2+ release.-Cao, X., Chen, J., Li, D., Xie, P., Xu, M., Lin, W., Li, S., Pan, G., Tang, Y., Xu, J., Olkkonen, V. M., Yan, D., Zhong, W. ORP4L couples IP3 to ITPR1 in control of endoplasmic reticulum calcium release.


Asunto(s)
Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Receptores de Esteroides/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Supervivencia Celular/fisiología , Citosol/metabolismo , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Células Jurkat , Mitocondrias/metabolismo , Fosforilación Oxidativa , Fosfolipasa C beta/metabolismo
7.
J Biol Chem ; 293(45): 17430-17441, 2018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30237164

RESUMEN

Phosphoinositide phospholipases C (PLCs) are a family of eukaryotic intracellular enzymes with important roles in signal transduction. In addition to their location at the plasma membrane, PLCs also exist within the cell nucleus where they are stored. We previously demonstrated that OSBP-related protein 4L (ORP4L) anchors cluster of differentiation 3ϵ (CD3ϵ) to the heterotrimeric G protein subunit (Gαq/11) to control PLCß3 relocation and activation. However, the underlying mechanism by which ORP4L facilitates PLCß3 translocation remains unknown. Here, using confocal immunofluorescence microscopy and coimmunoprecipitation assays, we report that ORP4L stimulates PLCß3 translocation from the nucleus to the plasma membrane in Jurkat T-cells in two steps. First, we found that ORP4L is required for the activation of Ras-related nuclear protein (RAN), a GTP-binding nuclear protein that binds to exportin 1 and eventually promotes the nuclear export of PLCß3. Second, we also observed that ORP4L interacts with vesicle-associated membrane protein-associated protein A (VAPA) through its two phenylalanines in an acidic tract (FFAT) motif. This complex enabled PLCß3 movement to the plasma membrane, indicating that PLCß3 translocation occurs in a VAPA-dependent manner. This study reveals detailed mechanistic insight into the role of ORP4L in PLCß3 redistribution from storage within the nucleus to the plasma membrane via RAN activation and interaction with VAPA in Jurkat T-cells.


Asunto(s)
Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Fosfolipasa C beta/metabolismo , Receptores de Esteroides/metabolismo , Linfocitos T/metabolismo , Transporte Activo de Núcleo Celular/fisiología , Membrana Celular/genética , Núcleo Celular/genética , Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Células HeLa , Células Hep G2 , Humanos , Células Jurkat , Células K562 , Fosfolipasa C beta/genética , Receptores de Esteroides/genética , Linfocitos T/citología , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Proteína de Unión al GTP ran/genética , Proteína de Unión al GTP ran/metabolismo
8.
Circ Res ; 119(12): 1296-1312, 2016 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-27729467

RESUMEN

RATIONALE: Macrophage survival within the arterial wall is a central factor contributing to atherogenesis. Oxysterols, major components of oxidized low-density lipoprotein, exert cytotoxic effects on macrophages. OBJECTIVE: To determine whether oxysterol-binding protein-related protein 4 L (ORP4L), an oxysterol-binding protein, affects macrophage survival and the pathogenesis of atherosclerosis. METHODS AND RESULTS: By hiring cell biological approaches and ORP4L-/- mice, we show that ORP4L coexpresses with and forms a complex with Gαq/11 and phospholipase C (PLC)-ß3 in macrophages. ORP4L facilitates G-protein-coupled ligand-induced PLCß3 activation, IP3 production, and Ca2+ release from the endoplasmic reticulum. Through this mechanism, ORP4L sustains antiapoptotic Bcl-XL expression through Ca2+-mediated c-AMP responsive element binding protein transcriptional regulation and thus protects macrophages from apoptosis. Excessive stimulation with the oxysterol 25-hydroxycholesterol disassembles the ORP4L/Gαq/11/PLCß3 complexes, resulting in reduced PLCß3 activity, IP3 production, and Ca2+ release, as well as decreased Bcl-XL expression and increased apoptosis. Overexpression of ORP4L counteracts these oxysterol-induced defects. Mice lacking ORP4L exhibit increased apoptosis of macrophages in atherosclerotic lesions and a reduced lesion size. CONCLUSIONS: ORP4L is crucial for macrophage survival. It counteracts the cytotoxicity of oxysterols/oxidized low-density lipoprotein to protect macrophage from apoptosis, thus playing an important role in the development of atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Aterosclerosis/prevención & control , Macrófagos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Esteroides/metabolismo , Transducción de Señal/fisiología , Animales , Aterosclerosis/patología , Supervivencia Celular/fisiología , Células Cultivadas , Humanos , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
9.
J Lipid Res ; 57(10): 1845-1853, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27530118

RESUMEN

Oxysterols are 27-carbon oxidized derivatives of cholesterol or by-products of cholesterol biosynthesis that can induce cell apoptosis in addition to a number of other bioactions. However, the mechanisms underlying this cytotoxicity are not completely understood. ORP8 is a member of the oxysterol binding protein-related protein (ORP) family, implicated in cellular lipid homeostasis, migration, and organization of the microtubule cytoskeleton. Here, we report that 25-hydroxycholesterol (OHC) induced apoptosis of the hepatoma cell lines, HepG2 and Huh7, via the endoplasmic reticulum (ER) stress response pathway, and ORP8 overexpression resulted in a similar cell response as 25-OHC, indicating a putative functional relationship between oxysterol cytotoxicity and ORP8. Further experiments demonstrated that ORP8 overexpression significantly enhanced the 25-OHC effect on ER stress and apoptosis in HepG2 cells. A truncated ORP8 construct lacking the ligand-binding domain or a closely related protein, ORP5, was devoid of this activity, evidencing for specificity of the observed effects. Importantly, ORP8 knockdown markedly dampened such responses to 25-OHC. Taken together, the present study suggests that ORP8 may mediate the cytotoxicity of 25-OHC.


Asunto(s)
Apoptosis/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Hidroxicolesteroles/toxicidad , Receptores de Esteroides/metabolismo , Apoptosis/genética , Estrés del Retículo Endoplásmico/genética , Células Hep G2 , Humanos , Receptores de Esteroides/genética
10.
J Biol Chem ; 290(14): 8876-87, 2015 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-25596532

RESUMEN

Human hepatoma (HCC) has been reported to be strongly resistant to Fas-mediated apoptosis. However, the underlying mechanisms are poorly understood. In this study the function of oxysterol-binding protein-related protein 8 (ORP8) in human hepatoma cells apoptosis was assessed. We found that ORP8 is down-regulated, whereas miR-143, which controls ORP8 expression, is up-regulated in clinical HCC tissues as compared with liver tissue from healthy subjects. ORP8 overexpression triggered apoptosis in primary HCC cells and cell lines, which coincided with a relocation of cytoplasmic Fas to the cell plasma membrane and FasL up-regulation. Co-culture of HepG2 cells or primary HCC cells with Jurkat T-cells or T-cells, respectively, provided further evidence that ORP8 increases HCC cell sensitivity to Fas-mediated apoptosis. ORP8-induced Fas translocation is p53-dependent, and FasL was induced upon ORP8 overexpression via the endoplasmic reticulum stress response. Moreover, ORP8 overexpression and miR-143 inhibition markedly inhibited tumor growth in a HepG2 cell xenograft model. These results indicate that ORP8 induces HCC cell apoptosis through the Fas/FasL pathway. The role of ORP8 in Fas translocation to the plasma membrane and its down-regulation by miR-143 offer a putative mechanistic explanation for HCC resistance to apoptosis. ORP8 may be a potential target for HCC therapy.


Asunto(s)
Apoptosis/fisiología , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Receptores de Esteroides/fisiología , Receptor fas/fisiología , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN , Citometría de Flujo , Humanos , Reacción en Cadena de la Polimerasa
11.
Exp Cell Res ; 322(2): 227-35, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24424245

RESUMEN

We earlier identified OSBP-related protein 8 (ORP8) as an endoplasmic reticulum/nuclear envelope oxysterol-binding protein implicated in cellular lipid homeostasis, migration, and organization of the microtubule cytoskeleton. Here, a yeast two-hybrid screen identified Homo sapiens sperm associated antigen 5 (SPAG5)/Astrin as interaction partner of ORP8. The putative interaction was further confirmed by pull-down and co-immunoprecipitation assays. ORP8 did not colocalize with kinetochore-associated SPAG5 in mitotic HepG2 or HuH7 cells, but overexpressed ORP8 was capable of recruiting SPAG5 onto endoplasmic reticulum membranes in interphase cells. In our experiments, 25-hydroxycholesterol (25OHC) retarded the HepG2 cell cycle, causing accumulation in G2/M phase; ORP8 overexpression resulted in the same phenotype. Importantly, ORP8 knock-down dramatically inhibited the oxysterol effect on HepG2 cell cycle, suggesting a mediating role of ORP8. Furthermore, knock-down of SPAG5 significantly reduced the effects of both ORP8 overexpression and 25OHC on the cell cycle, placing SPAG5 downstream of the two cell-cycle interfering factors. Taken together, the present results suggest that ORP8 may via SPAG5 mediate oxysterol interference of the HepG2 cell cycle.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Ciclo Celular/efectos de los fármacos , Hidroxicolesteroles/farmacología , Receptores de Esteroides/metabolismo , Western Blotting , Ciclo Celular/fisiología , Técnica del Anticuerpo Fluorescente , Células Hep G2 , Humanos , Inmunoprecipitación , Microscopía Fluorescente , Fracciones Subcelulares , Técnicas del Sistema de Dos Híbridos
12.
Exp Cell Res ; 318(15): 1933-45, 2012 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-22683860

RESUMEN

ORP8 is an oxysterol/cholesterol binding protein anchored to the endoplasmic reticulum and the nuclear envelope, and is abundantly expressed in the macrophage. We created and characterized mouse RAW264.7 macrophages with ORP8 stably silenced using shRNA lentiviruses. A microarray transcriptome and gene ontology pathway analysis revealed significant alterations in several nuclear pathways and ones associated with centrosome and microtubule organization. ORP8 knockdown resulted in increased expression and altered subcellular distribution of an interaction partner of ORP8, nucleoporin NUP62, with an intranuclear localization aspect and association with cytoplasmic vesicular structures and lamellipodial edges of the cells. Moreover, ORP8 silenced cells displayed enhanced migration, and a more pronounced microtubule cytoskeleton than controls expressing a non-targeting shRNA. ORP8 was shown to compete with Exo70 for interaction with NUP62, and NUP62 knockdown abolished the migration enhancement of ORP8-silenced cells, suggesting that the endogenous ORP8 suppresses migration via binding to NUP62. As a conclusion, the present study reveals new, unexpected aspects of ORP8 function in macrophages not directly involving lipid metabolism, but rather associated with nuclear functions, microtubule organization, and migration capacity.


Asunto(s)
Macrófagos/fisiología , Glicoproteínas de Membrana/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Esteroides/fisiología , Animales , Secuencia de Bases , Unión Competitiva , Ciclo Celular , Línea Celular , Movimiento Celular , Núcleo Celular/metabolismo , Proliferación Celular , Cartilla de ADN/genética , Técnicas de Silenciamiento del Gen , Macrófagos/citología , Glicoproteínas de Membrana/antagonistas & inhibidores , Glicoproteínas de Membrana/genética , Ratones , Microtúbulos/metabolismo , Proteínas de Complejo Poro Nuclear/antagonistas & inhibidores , Proteínas de Complejo Poro Nuclear/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Receptores de Esteroides/genética , Transcriptoma , Proteínas de Transporte Vesicular/metabolismo
13.
Biotechnol Appl Biochem ; 60(3): 330-5, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23621611

RESUMEN

The marine diatom Phaeodactylum tricornutum, a widely used forage species, has a storage lipid content of up to 30% dry cell weight. To explore the mechanism behind the high storage lipid accumulation in this diatom, acetyl-CoA carboxylase (ACCase), which catalyzes the first committed step of the fatty acid biosynthetic pathway, was characterized in this study. A homogeneous type of ACCase (PtACC) was identified from P. tricornutum by homology searches. The first exon of the ACCase gene (PtACC-1) was cloned. PtACC-1 was fused with a Myc epitope tag and cloned into plasmid pMD18 driven by the LacZ promoter and expressed in Escherichia coli. The expression of the PtACC-1-Myc protein was verified by Western blot. The neutral lipid content in transformed E. coli increased substantially by twofold as determined by Nile red fluorescent dye staining. Concomitantly, ACCase activity increased by 1.72-fold. The fatty acid composition, analyzed by GC-MS, demonstrated a significant difference in the ratio of saturated fatty acids and monounsaturated fatty acids (MUFAs). MUFAs of PtACC-1 expressing cells increased by 13%. This study represents the first characterization of the key domains of ACCase from a diatom and demonstrates high neutral lipid accumulation in E. coli expressing PtACC-1, providing an additional genetic resource with the potential for biodiesel development.


Asunto(s)
Diatomeas/genética , Escherichia coli/genética , Ácidos Grasos/metabolismo , Subunidades de Proteína/genética , Acetil-CoA Carboxilasa , Clonación Molecular/métodos , Diatomeas/metabolismo , Escherichia coli/metabolismo , Exones/genética , Ácidos Grasos/genética , Expresión Génica/genética , Filogenia , Estructura Terciaria de Proteína/genética , Subunidades de Proteína/metabolismo
14.
Circ Res ; 106(6): 1117-28, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20167927

RESUMEN

RATIONALE: AMP-activated protein kinase (AMPK) is an energy sensor and ubiquitously expressed in vascular cells. Recent studies suggest that AMPK activation improves endothelial function by counteracting oxidative stress in endothelial cells. How AMPK suppresses oxidative stress remains to be established. OBJECTIVE: The aim of this study is to examine the effects of AMPK in regulating NAD(P)H oxidase, oxidative stress, and endothelial function. METHODS AND RESULTS: The markers of oxidative stress, NAD(P)H oxidase subunit expression (gp91(phox), p47(phox), p67(phox), NOX1 to -4), NAD(P)H oxidase-mediated superoxide production, 26S proteasome activity, IkappaBalpha degradation, and nuclear translocation of nuclear factor (NF)-kappaB (p50 and p65) were examined in cultured human umbilical vein endothelial cells and mouse aortas isolated from AMPKalpha2 deficient mice. Compared to the wild type, acetylcholine-induced endothelium-dependent relaxation was significantly impaired in parallel with increased production of oxidants in AMPKalpha2(-/-) mice. Further, pretreatment of aorta with either superoxide dismutase (SOD) or tempol or apocynin significantly improved acetylcholine-induced endothelium-dependent relaxation in AMPKalpha2(-/-) mice. Analysis of aortic endothelial cells from AMPKalpha2(-/-) mice and human umbilical vein endothelial cells expressing dominant negative AMPK or AMPKalpha2-specific siRNA revealed that loss of AMPK activity increased NAD(P)H oxidase subunit expression (gp91(phox), p47(phox), p67(phox), NOX1 and -4), NAD(P)H oxidase-mediated superoxide production, 26S proteasome activity, IkappaBalpha degradation, and nuclear translocation of NF-kappaB (p50 and p65), whereas AMPK activation by AICAR or overexpression of constitutively active AMPK had the opposite effect. Consistently, we found that genetic deletion of AMPKalpha2 in low-density lipoprotein receptor knockout (LDLr(-/-)) strain markedly increased 26S proteasome activity, IkappaB degradation, NF-kappaB transactivation, NAD(P)H oxidase subunit overexpression, oxidative stress, and endothelial dysfunction, all of which were largely suppressed by chronic administration of MG132, a potent cell permeable proteasome inhibitor. CONCLUSIONS: We conclude that AMPKalpha2 functions as a physiological suppressor of NAD(P)H oxidase and ROS production in endothelial cells. In this way, AMPK maintains the nonatherogenic and noninflammatory phenotype of endothelial cells.


Asunto(s)
Proteínas Quinasas Activadas por AMP/deficiencia , Células Endoteliales/enzimología , Endotelio Vascular/enzimología , NADPH Oxidasas/metabolismo , Estrés Oxidativo , Complejo de la Endopetidasa Proteasomal/metabolismo , Vasodilatación , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Proteínas Quinasas Activadas por AMP/genética , Acetofenonas/farmacología , Acetilcolina/farmacología , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Antioxidantes/farmacología , Células Cultivadas , Óxidos N-Cíclicos/farmacología , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Eliminación de Gen , Regulación Enzimológica de la Expresión Génica , Humanos , Proteínas I-kappa B/metabolismo , Mediadores de Inflamación/sangre , Interferón gamma/sangre , Interleucina-2/sangre , Isoenzimas , Leupeptinas/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/genética , Inhibidor NF-kappaB alfa , FN-kappa B/metabolismo , Nitroprusiato/farmacología , Estrés Oxidativo/efectos de los fármacos , Inhibidores de Proteasoma , Interferencia de ARN , Receptores de LDL/deficiencia , Receptores de LDL/genética , Ribonucleótidos/farmacología , Marcadores de Spin , Superóxido Dismutasa/metabolismo , Superóxidos/metabolismo , Transfección , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
15.
Exp Cell Res ; 317(16): 2353-63, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21669198

RESUMEN

ORP7 is a member of oxysterol-binding protein (OSBP) family, the function of which has remained obscure. In this study, we identified by yeast two-hybrid screening an interaction partner of ORP7, GATE-16, which (i) regulates Golgi SNARE of 28kDa (GS28) function and stability, and (ii) plays a role in autophagosome biogenesis. The interaction was confirmed by bimolecular fluorescence complementation (BiFC) assay in living cells. The interacting regions were delineated within aa 1-142 of ORP7 and aa 30-117 of GATE-16. ORP7 knock-down in 293A cells resulted in a 40% increase of GS28 protein while ORP7 overexpression had the opposite effect (25% decrease of GS28). We show evidence that the regulation of GS28 by ORP7 does not occur at the level of transcription, but involves degradation of GS28 on proteasomes. Truncated ORP7 that lacks the GATE-16 binding region failed to affect GS28 stability, evidencing for specificity of the observed effect. Similar to ORP7 overexpression, treatment of cells with 25-hydroxycholesterol (25-OH) resulted in GS28 destabilization, which was potentiated by excess ORP7 and inhibited by ORP7 silencing. Overexpression of ORP7 led in most cells to formation of vacuolar structures positive for RFP-LC3, thus representing autophagic elements. Also GATE-16 was found in the vacuolar ORP7-positive elements, suggesting that excess ORP7 increases entrapment of GATE-16 in autophagosomes. Taken together, our results suggest that ORP7 negatively regulates GS28 protein stability via sequestration of GATE-16, and may mediate the effect of 25-OH on GS28 and Golgi function.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Microfilamentos/metabolismo , Proteínas Qb-SNARE/metabolismo , Receptores de Esteroides/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Familia de las Proteínas 8 Relacionadas con la Autofagia , Sitios de Unión/fisiología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Citoplasma/metabolismo , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Aparato de Golgi/metabolismo , Células HEK293 , Humanos , Hidroxicolesteroles/farmacología , Leupeptinas/farmacología , Proteínas de Microfilamentos/genética , Fagosomas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores de Proteasoma , Unión Proteica/fisiología , Dominios y Motivos de Interacción de Proteínas/fisiología , Mapeo de Interacción de Proteínas/métodos , Proteolisis/efectos de los fármacos , Proteínas Qb-SNARE/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Receptores de Esteroides/antagonistas & inhibidores , Receptores de Esteroides/genética , Eliminación de Secuencia/fisiología , Transfección , Técnicas del Sistema de Dos Híbridos
16.
Front Pharmacol ; 13: 811406, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35211017

RESUMEN

Anthracyclines, such as doxorubicin, represent one group of chemotherapy drugs with the most cardiotoxicity. Despite that anthracyclines are capable of treating assorted solid tumors and hematological malignancies, the side effect of inducing cardiac dysfunction has hampered their clinical use. Currently, the mechanism underlying anthracycline cardiotoxicity remains obscure. Increasing evidence points to mitochondria, the energy factory of cardiomyocytes, as a major target of anthracyclines. In this review, we will summarize recent findings about mitochondrial mechanism during anthracycline cardiotoxicity. In particular, we will focus on the following aspects: 1) the traditional view about anthracycline-induced reactive oxygen species (ROS), which is produced by mitochondria, but in turn causes mitochondrial injury. 2) Mitochondrial iron-overload and ferroptosis during anthracycline cardiotoxicity. 3) Autophagy, mitophagy and mitochondrial dynamics during anthracycline cardiotoxicity. 4) Anthracycline-induced disruption of cardiac metabolism.

17.
Nat Commun ; 13(1): 4390, 2022 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-35906240

RESUMEN

Lipid remodeling is crucial for malignant cell transformation and tumorigenesis, but the precise molecular processes involved and direct evidences for these in vivo remain elusive. Here, we report that oxysterol-binding protein (OSBP)-related protein 4 L (ORP4L) is expressed in adult T-cell leukemia (ATL) cells but not normal T-cells. In ORP4L knock-in T-cells, ORP4L dimerizes with OSBP to control the shuttling of OSBP between the Golgi apparatus and the plasma membrane (PM) as an exchanger of phosphatidylinositol 4-phosphate [PI(4)P]/cholesterol. The PI(4)P arriving at the PM via this transport machinery replenishes phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] and phosphatidylinositol (3,4,5) trisphosphate [PI(3,4,5)P3] biosynthesis, thus contributing to PI3K/AKT hyperactivation and T-cell deterioration in vitro and in vivo. Disruption of ORP4L and OSBP dimerization disables PI(4)P transport and T-cell leukemogenesis. In summary, we identify a non-vesicular lipid transport machinery between Golgi and PM maintaining the oncogenic signaling competence initiating T-cell deterioration and leukemogenesis.


Asunto(s)
Fosfatidilinositol 3-Quinasas , Receptores de Esteroides , Carcinogénesis , Humanos , Fosfatidilinositol 4,5-Difosfato , Fosfatos de Fosfatidilinositol/metabolismo , Fosfatidilinositoles , Receptores de Esteroides/metabolismo , Linfocitos T/metabolismo
18.
Sci Transl Med ; 14(629): eabh2548, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-35080912

RESUMEN

Leukemia stem cells (LSCs) propagate leukemia and are responsible for the high frequency of relapse of treated patients. The ability to target LSCs remains elusive, indicating a need to understand the underlying mechanism of LSC formation. Here, we report that miR-31-5p is reduced or undetectable in human LSCs compared to hematopoietic stem progenitor cells (HSPCs). Inhibition of miR-31-5p in HSPCs promotes the expression of its target gene FIH, encoding FIH [factor inhibiting hypoxia-inducing factor 1α (HIF-1α)], to suppress HIF-1α signaling. Increased FIH resulted in a switch from glycolysis to oxidative phosphorylation (OXPHOS) as the predominant mode of energy metabolism and increased the abundance of the oncometabolite fumarate. Increased fumarate promoted the conversion of HSPCs to LSCs and initiated myeloid leukemia-like disease in NOD-Prkdcscid IL2rgtm1/Bcgen (B-NDG) mice. We further demonstrated that miR-31-5p inhibited long- and short-term hematopoietic stem cells with a high frequency of LSCs. In combination with the chemotherapeutic agent Ara-C (cytosine arabinoside), restoration of miR-31-5p using G7 poly (amidoamine) nanosized dendriplex encapsulating miR-31-5p eliminated LSCs and inhibited acute myeloid leukemia (AML) progression in patient-derived xenograft mouse models. These results demonstrated a mechanism of HSC malignant transformation through altered energy metabolism and provided a potential therapeutic strategy to treat patients with AML.


Asunto(s)
Leucemia Mieloide Aguda , MicroARNs , Animales , Fumaratos , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Ratones , Ratones Endogámicos NOD , MicroARNs/genética , MicroARNs/metabolismo , Células Madre Neoplásicas/patología
19.
Exp Cell Res ; 316(19): 3304-16, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-20599956

RESUMEN

We characterize here ORP11, a member of the oxysterol-binding protein family. ORP11 is present at highest levels in human ovary, testis, kidney, liver, stomach, brain, and adipose tissue. Immunohistochemistry demonstrates abundant ORP11 in the epithelial cells of kidney tubules, testicular tubules, caecum, and skin. ORP11 in HEK293 cells resides on Golgi complex and LE, co-localizing with GFP-Rab9, TGN46, GFP-Rab7, and a fluorescent medial-trans-Golgi marker. Under electron microscopic observation, cells overexpressing ORP11 displayed lamellar lipid bodies associated with vacuolar structures or the Golgi complex, indicating a disturbance of lipid trafficking. N-terminal fragment of ORP11 (aa 1-292) localized partially to Golgi, but displayed enhanced localization on Rab7- and Rab9-positive LE, while the C-terminal ligand-binding domain (aa 273-747) was cytosolic, demonstrating that the membrane targeting determinants are N-terminal. Yeast two-hybrid screen revealed interaction of ORP11 with the related ORP9. The interacting region was delineated within aa 98-372 of ORP9 and aa 154-292 of ORP11. Overexpressed ORP9 was able to recruit EGFP-ORP11 to membranes, and ORP9 silencing inhibited ORP11 Golgi association. The results identify ORP11 as an OSBP homologue distributing at the Golgi-LE interface and define the ORP9-ORP11 dimer as a functional unit that may act as an intracellular lipid sensor or transporter.


Asunto(s)
Endosomas/metabolismo , Aparato de Golgi/metabolismo , Multimerización de Proteína , Receptores de Esteroides/metabolismo , Sitios de Unión , Western Blotting , Línea Celular , Endosomas/ultraestructura , Silenciador del Gen , Aparato de Golgi/ultraestructura , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Inmunohistoquímica , Lípidos/química , Especificidad de Órganos , Fosfatidilinositoles/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Transporte de Proteínas , Receptores de Esteroides/química , Fracciones Subcelulares/metabolismo , Vacuolas/metabolismo , Vacuolas/ultraestructura
20.
J Med Chem ; 64(4): 2010-2023, 2021 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-33543615

RESUMEN

Hsp90 is a new promising target for cancer treatment. Many inhibitors have been discovered as therapeutic agents, and some have passed Phase I and II. However, no one is approved by FDA yet. Novel and druggable Hsp90 inhibitors are still demanding. Here, we report a new way to discover high potent Hsp90 inhibitors as antinasopharyngeal carcinoma agents through assembling fragments. With chemotyping analysis, we extract seven chemotypes from 3482 known Hsp90 inhibitors, screen 500 fragments that are compatible with the chemotypes, and confirm 15 anti-Hsp90 fragments. Click chemistry is employed to construct 172 molecules and synthesize 21 compounds among them. The best inhibitor 3d was further optimized and resulted in more potent 4f (IC50 = 0.16 µM). In vitro and in vivo experiments confirmed that 4f is a promising agent against nasopharyngeal carcinoma. This study may provide a strategy in discovering new ligands against targets without well-understood structures.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Carcinoma Nasofaríngeo/tratamiento farmacológico , Neoplasias Nasofaríngeas/tratamiento farmacológico , Triazoles/uso terapéutico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/metabolismo , Línea Celular Tumoral , Bases de Datos de Compuestos Químicos , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Masculino , Ratones Desnudos , Simulación de Dinámica Molecular , Unión Proteica , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/metabolismo , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Triazoles/síntesis química , Triazoles/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA