Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Br J Cancer ; 122(6): 904-911, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32001831

RESUMEN

BACKGROUND: Nicotine is a major tobacco component and found at circulating concentrations in smokers' bloodstreams. Although considered a non-carcinogenic substance, nicotine rapidly defuses to tissues after being inhaled, inviting effects on cellular physiology, particularly in the lung. Widespread increased use of nicotine-based e-cigarettes, especially in younger adults, creates an urgent need for improved understanding of nicotine's potential to impact human health. METHODS: Biological and biochemistry methods were used to interrogate the potential for nicotine to weaken the genetic integrity of murine and human-lung epithelial cells. RESULTS: We demonstrate that nicotine potentiates the growth of the lung epithelial cells in a dose-response fashion. Nicotine elicits an acute increase in reactive oxygen species (ROS), which persists at moderately high levels throughout the duration of nicotine exposure. The aberrant increases in ROS appear to induce ER stress and UPR activation, as reflected by BIP upregulation and PERK phosphorylation. Furthermore, prolonged nicotine exposure interferes with p53 function triggered by sodium arsenite. Unless p53 is suppressed, persistent nicotine exposure does not induce colony formation by lung epithelial cells in soft agar. CONCLUSION: The data suggest that nicotine treatment, by perturbing intracellular redox state and altering p53 function, can create a pro-tumorigenic environment in lung epithelium. The results suggest caution in using nicotine replacement therapies and e-cigarettes.


Asunto(s)
Carcinogénesis/patología , Células Epiteliales/patología , Neoplasias Pulmonares/etiología , Nicotina/efectos adversos , Animales , Humanos , Neoplasias Pulmonares/patología , Ratones
2.
J Pathol ; 232(3): 369-81, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24421076

RESUMEN

Although the incidence of breast cancer metastasis (BCM) in brain has increased significantly in triple-negative breast cancer (TNBC), the mechanisms remain elusive. Using in vivo mouse models for BCM in brain, we observed that TNBC cells crossed the blood-brain barrier (BBB), lodged in the brain microvasculature and remained adjacent to brain microvascular endothelial cells (BMECs). Breaching of the BBB in vivo by TNBCs resulted in increased BBB permeability and changes in ZO-1 and claudin-5 tight junction (TJ) protein structures. Angiopoietin-2 expression was elevated in BMECs and was correlated with BBB disruption. Secreted Ang-2 impaired TJ structures and increased BBB permeability. Treatment of mice with the neutralizing Ang-2 peptibody trebananib prevented changes in the BBB integrity and BMEC destabilization, resulting in inhibition of TNBC colonization in brain. Thus, Ang-2 is involved in initial steps of brain metastasis cascade, and inhibitors for Ang-2 may serve as potential therapeutics for brain metastasis.


Asunto(s)
Angiopoyetina 2/metabolismo , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundario , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Barrera Hematoencefálica/patología , Permeabilidad Capilar/fisiología , Femenino , Xenoinjertos , Humanos , Inmunohistoquímica , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Uniones Estrechas/metabolismo , Uniones Estrechas/patología , Neoplasias de la Mama Triple Negativas/patología
3.
Int J Cancer ; 134(5): 1034-44, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23934616

RESUMEN

Neuropeptide substance P (SP) has been implicated in inflammation, pain, depression and breast cancer cell (BCC) growth. Here, we examined the role of SP in trafficking of BCCs (human MDA-MB-231 and MDA-MB-231BrM2 cells) across the blood-brain barrier (BBB) and brain microvascular endothelial cells (BMECs) using in vitro and in vivo models. SP was secreted from BCCs and mediated adhesion and transmigration of BCCs across human BMECs (HBMECs) in vitro. SP induced activation of HBMECs, leading to secretion of Tumor Necrosis Factor alpha (TNF-α) and angiopoietin-2 (Ang-2) from HBMECs, resulting in changes in localization and distribution of tight junction (TJ) ZO-1 (tight junction protein zonula occludins-1) and claudin-5 structures as well as increased permeability of HBMECs. Using spontaneous breast cancer metastasis mouse model (syngeneic) of GFP-4T1-BrM5 mammary tumor cells administered into mammary fat pads of Balb/c mice, SP inhibitor spantide III inhibited in vivo changes in permeability of the BBB and BMEC-TJs ZO-1 and claudin-5 structures as well as decreased tumor cell colonization in brain. Thus, SP secreted from BCCs induces transmigration of BCCs across the BBB, leading to activation of BMECs and secretion of TNF-α and Ang-2, resulting in BBB impairment and colonization of tumor cells in brain. Therefore, therapies based on SP inhibition in combination with other therapies may prevent breaching of the BBB by BCCs and their colonization in brain.


Asunto(s)
Barrera Hematoencefálica , Neoplasias de la Mama/patología , Células Endoteliales/fisiología , Sustancia P/fisiología , Uniones Estrechas/fisiología , Animales , Adhesión Celular , Movimiento Celular , Citoesqueleto/química , Femenino , Humanos , Ratones , Microvasos/citología , Permeabilidad , Sustancia P/análogos & derivados , Sustancia P/antagonistas & inhibidores , Sustancia P/farmacología , Factor de Necrosis Tumoral alfa/metabolismo
4.
Blood ; 117(3): 827-38, 2011 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-21063029

RESUMEN

Endocannabinoids are arachidonic acid derivatives and part of a novel bioactive lipid signaling system, along with their G-coupled cannabinoid receptors (CB1 and CB2) and the enzymes involved in their biosynthesis and degradation. However, their roles in hematopoiesis and hematopoietic stem and progenitor cell (HSPC) functions are not well characterized. Here, we show that bone marrow stromal cells express endocannabinoids (anandamide and 2-arachidonylglycerol), whereas CB2 receptors are expressed in human and murine HSPCs. On ligand stimulation with CB2 agonists, CB2 receptors induced chemotaxis, migration, and enhanced colony formation of bone marrow cells, which were mediated via ERK, PI3-kinase, and Gαi-Rac1 pathways. In vivo, the CB2 agonist AM1241 induced mobilization of murine HSPCs with short- and long-term repopulating abilities. In addition, granulocyte colony-stimulating factor -induced mobilization of HSPCs was significantly decreased by specific CB2 antagonists and was impaired in Cnr2(-/-) cannabinoid type 2 receptor knockout mice. Taken together, these results demonstrate that the endocannabinoid system is involved in hematopoiesis and that CB2/CB2 agonist axis mediates repopulation of hematopoiesis and mobilization of HSPCs. Thus, CB2 agonists may be therapeutically applied in clinical conditions, such as bone marrow transplantation.


Asunto(s)
Hematopoyesis/fisiología , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/fisiología , Receptor Cannabinoide CB2/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Moduladores de Receptores de Cannabinoides/metabolismo , Cannabinoides/farmacología , Movimiento Celular/efectos de los fármacos , Ciclohexanoles/farmacología , Femenino , Citometría de Flujo , Hematopoyesis/efectos de los fármacos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Humanos , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/genética , Células del Estroma/metabolismo
5.
Sci Rep ; 12(1): 5328, 2022 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-35351947

RESUMEN

While the prevalence of breast cancer metastasis in the brain is significantly higher in triple negative breast cancers (TNBCs), there is a lack of novel and/or improved therapies for these patients. Monoacylglycerol lipase (MAGL) is a hydrolase involved in lipid metabolism that catalyzes the degradation of 2-arachidonoylglycerol (2-AG) linked to generation of pro- and anti-inflammatory molecules. Here, we targeted MAGL in TNBCs, using a potent carbamate-based inhibitor AM9928 (hMAGL IC50 = 9 nM) with prolonged pharmacodynamic effects (46 h of target residence time). AM9928 blocked TNBC cell adhesion and transmigration across human brain microvascular endothelial cells (HBMECs) in 3D co-cultures. In addition, AM9928 inhibited the secretion of IL-6, IL-8, and VEGF-A from TNBC cells. TNBC-derived exosomes activated HBMECs resulting in secretion of elevated levels of IL-8 and VEGF, which were inhibited by AM9928. Using in vivo studies of syngeneic GFP-4T1-BrM5 mammary tumor cells, AM9928 inhibited tumor growth in the mammary fat pads and attenuated blood brain barrier (BBB) permeability changes, resulting in reduced TNBC colonization in brain. Together, these results support the potential clinical application of MAGL inhibitors as novel treatments for TNBC.


Asunto(s)
Monoacilglicerol Lipasas , Neoplasias de la Mama Triple Negativas , Encéfalo/metabolismo , Células Endoteliales/metabolismo , Humanos , Inflamación , Monoacilglicerol Lipasas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
6.
Sci Rep ; 12(1): 22255, 2022 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-36564457

RESUMEN

Triple-negative breast cancer (TNBC) is associated with high mortality due to the high expression of pro-inflammatory cytokines and lack of targeted therapies. N-acylethanolamine acid amidase (NAAA) is an N-terminal cysteine hydrolase that promotes inflammatory responses through the deactivation of Palmitoylethanolamide (PEA), an endogenous bioactive lipid mediator. Here, we examined NAAA expression in TNBC cells (MDA-MB-231 and MDA-MB-BrM2 cells) and the effects of NAAA inhibition on TNBC tumor growth, using a selective NAAA inhibitor AM11095 (IC50 = 20 nM). TNBC cells expressed elevated levels of full-length and splice mRNAs naaa variants. TNBC cells also express the N-acyl ethanol amides and elevated levels of the two fatty acid cores arachidonic (AA) and docosahexaenoic (DHA). PEA or AM11095 inhibited the secretion of IL-6 and IL-8, reduced the activation of the NF-kB pathway, decreased the expression of VEGF and Placental growth factor (PLGF) in TNBCs, and inhibited tumor cell migration in vitro. Using cellular magnetic resonance imaging (MRI), body images of mice administered with human MDA-MB-BrM2 cells treated with AM11095 showed a significant decrease in tumor numbers with a lower volume of tumors and increased mice survival. Mice untreated or treated with vehicle control showed a high number of tumors with high volumes in multiple organs. Thus, NAAA inhibition may constitute a potential therapeutic approach in the management of TNBC-associated inflammation and tumor growth.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Ratones , Humanos , Femenino , Animales , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Factor de Crecimiento Placentario/uso terapéutico , Inflamación/tratamiento farmacológico , Amidas/uso terapéutico
7.
J Biol Chem ; 285(34): 26190-8, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20511222

RESUMEN

Reactive molecules have diverse effects on cells and contribute to several pathological conditions. Cells have evolved complex protective systems to neutralize these molecules and restore redox homeostasis. Previously, we showed that association of nuclear factor (NF)-erythroid-derived 2 (E2)-related factor 2 (NRF2) with the nuclear matrix protein NRP/B was essential for the transcriptional activity of NRF2 target genes in tumor cells. The present study demonstrates the molecular mechanism by which NRP/B, via NRF2, modulates the transcriptional activity of antioxidant response element (ARE)-driven genes. NRP/B is localized in the nucleus of primary brain tissue and human neuroblastoma (SH-SY5Y) cells. Treatment with hydrogen peroxide (H(2)O(2)) enhances the nuclear colocalization of NRF2 and NRP/B and induces heme oxygenase 1 (HO1). Treatment of NRP/B or NRF2 knockdowns with H(2)O(2) induced apoptosis. Co-expression of NRF2 with members of the Kelch protein family, NRP/B, MAYVEN, or MAYVEN-related protein 2 (MRP2), revealed that the NRF2-NRP/B complex is important for the transcriptional activity of ARE-driven genes HO1 and NAD(P)H:quinine oxidoreductase 1 (NQO1). NRP/B interaction with Nrf2 was mapped to NRF2 ECH homology 4 (Neh4)/Neh5 regions of NRF2. NRP/B mutations that resulted in low binding affinity to NRF2 were unable to activate NRF2-modulated transcriptional activity of the ARE-driven genes, HO1 and NQO1. Thus, the interaction of NRP/B with the Neh4/Neh5 domains of NRF2 is indispensable for activation of NRF2-mediated ARE-driven antioxidant and detoxifying genes that confer cellular defense against oxidative stress-induced damage.


Asunto(s)
Proteínas de Microfilamentos/fisiología , Factor 2 Relacionado con NF-E2/metabolismo , Neuropéptidos/fisiología , Proteínas Asociadas a Matriz Nuclear/fisiología , Proteínas Nucleares/fisiología , Estrés Oxidativo/genética , Activación Transcripcional , Antioxidantes , Química Encefálica , Línea Celular Tumoral , Regulación de la Expresión Génica , Humanos , Neuroblastoma/química , Oxidación-Reducción , Elementos de Respuesta
8.
J Biol Chem ; 285(46): 35471-8, 2010 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-20826813

RESUMEN

Endocannabinoids are lipid signaling molecules that act via G-coupled receptors, CB(1) and CB(2). The endocannabinoid system is capable of activation of distinct signaling pathways on demand in response to pathogenic events or stimuli, hereby enhancing cell survival and promoting tissue repair. However, the role of endocannabinoids in hematopoietic stem and progenitor cells (HSPCs) and their interaction with hematopoietic stem cells (HSC) niches is not known. HSPCs are maintained in the quiescent state in bone marrow (BM) niches by intrinsic and extrinsic signaling. We report that HSPCs express the CB(1) receptors and that BM stromal cells secrete endocannabinoids, anandamide (AEA) (35 pg/10(7) cells), and 2-AG (75.2 ng/10(7) cells). In response to the endotoxin lipopolysaccharide (LPS), elevated levels of AEA (75.6 pg/10(7) cells) and 2-AG (98.8 ng/10(7) cells) were secreted from BM stromal cells, resulting in migration and trafficking of HSPCs from the BM niches to the peripheral blood. Furthermore, administration of exogenous cannabinoid CB(1) agonists in vivo induced chemotaxis, migration, and mobilization of human and murine HSPCs. Cannabinoid receptor knock-out mice Cnr1(-/-) showed a decrease in side population (SP) cells, whereas fatty acid amide hydrolase (FAAH)(-/-) mice, which have elevated levels of AEA, yielded increased colony formation as compared with WT mice. In addition, G-CSF-induced mobilization in vivo was modulated by endocannabinoids and was inhibited by specific cannabinoid antagonists as well as impaired in cannabinoid receptor knock-out mice Cnr1(-/-), as compared with WT mice. Thus, we propose a novel function of the endocannabinoid system, as a regulator of HSPC interactions with their BM niches, where endocannabinoids are expressed in HSC niches and under stress conditions, endocannabinoid expression levels are enhanced to induce HSPC migration for proper hematopoiesis.


Asunto(s)
Células de la Médula Ósea/metabolismo , Moduladores de Receptores de Cannabinoides/biosíntesis , Endocannabinoides , Células Madre Hematopoyéticas/metabolismo , Células del Estroma/metabolismo , Amidohidrolasas/genética , Amidohidrolasas/metabolismo , Animales , Ácidos Araquidónicos/biosíntesis , Western Blotting , Células de la Médula Ósea/citología , Moduladores de Receptores de Cannabinoides/fisiología , Comunicación Celular/fisiología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Ciclohexanoles/farmacología , Femenino , Citometría de Flujo , Glicéridos/biosíntesis , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/citología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Alcamidas Poliinsaturadas , Receptor Cannabinoide CB1/agonistas , Receptor Cannabinoide CB1/genética , Receptor Cannabinoide CB1/metabolismo , Receptor Cannabinoide CB2/agonistas , Receptor Cannabinoide CB2/genética , Receptor Cannabinoide CB2/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células de Población Lateral/citología , Células de Población Lateral/metabolismo , Nicho de Células Madre/citología , Nicho de Células Madre/metabolismo , Células del Estroma/citología
9.
Transfusion ; 51 Suppl 4: 65S-71S, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22074629

RESUMEN

The cannabinoid receptors CB(1) and CB(2) are seven-transmembrane Gαi protein-coupled receptors and are expressed in certain mature hematopoietic cells. We recently showed that these receptors are expressed in murine and human hematopoietic stem cells (HSCs) and that CB(2) agonists induced chemotaxis, enhanced colony formation of marrow cells, as well as caused in vivo mobilization of murine HSCs with short- and long-term repopulating abilities. Based on these observations, we have further explored the role of CB(2) and its agonist AM1241 on hematopoietic recovery following sublethal irradiation in mice. Cannabinoid receptor 2 knockout mice (Cnr2(-/-) deficient mice) exhibited impaired recovery following sublethal irradiation as compared with irradiated wild-type (WT) mice, as determined by low colony-forming units and low peripheral blood counts. WT mice treated with CB(2) agonist AM1241 following sublethal irradiation demonstrated accelerated marrow recovery and increased total marrow cells (approximately twofold) and total lineage- c-kit(+) cells (approximately sevenfold) as well as enhanced HSC survival as compared with vehicle control-treated mice. When the CB(2) agonist AM1241 was administered to WT mice 12 days before their sublethal irradiation, analysis of hematopoiesis in these mice showed decreased apoptosis of HSCs, enhanced survival of HSCs, as well as increase in total marrow cells and c-kit+ cells in the marrow. Thus, CB(2) agonist AM1241 promoted recovery after sublethal irradiation by inhibiting apoptosis of HSCs and promoting survival, as well as enhancing the number of HSCs entering the cell cycle.


Asunto(s)
Moduladores de Receptores de Cannabinoides/fisiología , Endocannabinoides , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Receptor Cannabinoide CB1/fisiología , Receptor Cannabinoide CB2/fisiología , Animales , Apoptosis , Cannabinoides/farmacología , Ciclo Celular , Movimiento Celular/fisiología , Ensayo de Unidades Formadoras de Colonias , Hematopoyesis/efectos de la radiación , Humanos , Ratones , Receptor Cannabinoide CB2/antagonistas & inhibidores , Nicho de Células Madre , Irradiación Corporal Total
10.
Biochim Biophys Acta ; 1788(5): 1159-67, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19366584

RESUMEN

Little direct information is available regarding the influence of membrane environment on transmembrane (TM) G-protein-coupled receptor (GPCR) conformation and dynamics. The human CB1 cannabinoid receptor (hCB1) is a prominent GPCR pharmacotherapeutic target in which helix 7 appears critical to ligand recognition. We have chemically synthesized a hCB1 peptide corresponding to a segment of TM helix 7 and the entire contiguous helix 8 domain (fourth cytoplasmic loop) and reconstituted it in defined phospholipid-bilayer model membranes. Using an NMR-based strategy combined with molecular dynamics simulations, we provide the first direct experimental description of the orientation of hCB1 helix 7 in phospholipid membranes of varying thickness and the mechanism by which helix-7 conformation adjusts to avoid hydrophobic mismatch. Solid-state (15)N NMR data show that hCB1 helices 7 and 8 reconstituted into phospholipid bilayers are oriented in a TM and in-plane (i.e., parallel to the phospholipid membrane surface) fashion, respectively. TM helix orientation is influenced by the thickness of the hydrophobic membrane bilayer as well as the interaction of helix 8 with phospholipid polar headgroups. Molecular dynamics simulations show that a decrease in phospholipid chain-length induces a kink at P394 in TM helix 7 to avoid hydrophobic mismatch. Thus, the NP(X)nY motif found in hCB1 and highly conserved throughout the GPCR superfamily is important for flexing helix 7 to accommodate bilayer thickness. Dynamic modulation of hCB1-receptor TM helix conformation by its membrane environment may have general relevance to GPCR structure and function.


Asunto(s)
Receptor Cannabinoide CB1/química , Secuencia de Aminoácidos , Fenómenos Biofísicos , Dicroismo Circular , Dimiristoilfosfatidilcolina/química , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Técnicas In Vitro , Membrana Dobles de Lípidos/química , Modelos Moleculares , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Fosfatidilcolinas/química , Estructura Secundaria de Proteína , Receptor Cannabinoide CB1/genética , Termodinámica
11.
J Immunol ; 181(9): 6406-16, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18941231

RESUMEN

HIV-1 infection has significant effect on the immune system as well as on the nervous system. Breakdown of the blood-brain barrier (BBB) is frequently observed in patients with HIV-associated dementia (HAD) despite lack of productive infection of human brain microvascular endothelial cells (HBMEC). Cellular products and viral proteins secreted by HIV-1 infected cells, such as the HIV-1 Gp120 envelope glycoprotein, play important roles in BBB impairment and HIV-associated dementia development. HBMEC are a major component of the BBB. Using cocultures of HBMEC and human astrocytes as a model system for human BBB as well as in vivo model, we show for the first time that cannabinoid agonists inhibited HIV-1 Gp120-induced calcium influx mediated by substance P and significantly decreased the permeability of HBMEC as well as prevented tight junction protein down-regulation of ZO-1, claudin-5, and JAM-1 in HBMEC. Furthermore, cannabinoid agonists inhibited the transmigration of human monocytes across the BBB and blocked the BBB permeability in vivo. These results demonstrate that cannabinoid agonists are able to restore the integrity of HBMEC and the BBB following insults by HIV-1 Gp120. These studies may lead to better strategies for treatment modalities targeted to the BBB following HIV-1 infection of the brain based on cannabinoid pharmacotherapies.


Asunto(s)
Fármacos Anti-VIH/farmacología , Ácidos Araquidónicos/farmacología , Encéfalo/irrigación sanguínea , Encéfalo/virología , Moduladores de Receptores de Cannabinoides/farmacología , Endotelio Vascular/virología , Glicéridos/farmacología , Proteína gp120 de Envoltorio del VIH/antagonistas & inhibidores , VIH-1/efectos de los fármacos , Complejo SIDA Demencia/enzimología , Complejo SIDA Demencia/patología , Complejo SIDA Demencia/prevención & control , Complejo SIDA Demencia/virología , Amidohidrolasas/antagonistas & inhibidores , Fármacos Anti-VIH/uso terapéutico , Ácidos Araquidónicos/fisiología , Benzamidas/farmacología , Benzamidas/uso terapéutico , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Moduladores de Receptores de Cannabinoides/agonistas , Moduladores de Receptores de Cannabinoides/fisiología , Carbamatos/farmacología , Carbamatos/uso terapéutico , Línea Celular , Técnicas de Cocultivo , Endocannabinoides , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/enzimología , Endotelio Vascular/fisiología , Glicéridos/fisiología , Proteína gp120 de Envoltorio del VIH/fisiología , VIH-1/fisiología , Humanos , Microcirculación/efectos de los fármacos , Microcirculación/fisiología , Receptor Cannabinoide CB1/fisiología
12.
Mol Cell Biol ; 26(22): 8371-84, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16982692

RESUMEN

The actin-based cytoskeleton is essential for the generation and maintenance of cell polarity, cellular motility, and the formation of neural cell processes. MRP2 is an actin-binding protein of the kelch-related protein family. While MRP2 has been shown to be expressed specifically in brain, its function is still unknown. Here, we report that in neuronal growth factor (NGF)-induced PC12 cells, MRP2 was expressed along the neurite processes and colocalized with Talin at the growth cones. MRP2 mRNA and protein levels were up-regulated in PC12 cells following NGF stimulation. Moreover, treatment of PC12 cells with interfering RNAs for MRP2 and glycogen synthase kinase 3beta (GSK3beta) resulted in the inhibition of neurite outgrowth. A significant decrease in MRP2 expression levels was observed following GSK3beta inhibition, which was correlated with the inhibited neurite outgrowth, while GSK3beta overexpression was found to increase MRP2 expression levels. MRP2 interacted with GSK3beta through its NH2 terminus containing the BTB domain, and these molecules colocalized along neurite processes and growth cones in differentiated PC12 cells and rat primary hippocampal neurons. Additionally, increased associations of MRP2 with GSK3beta and MRP2 with actin were observed in the NGF-treated PC12 cells. Thus, this study provides, for the first time, insights into the involvement of MRP2 in neurite outgrowth, which occurs in a GSK3beta-dependent manner.


Asunto(s)
Glucógeno Sintasa Quinasa 3/fisiología , Proteínas de Microfilamentos/fisiología , Factor de Crecimiento Nervioso/farmacología , Neuritas/fisiología , Neuronas/fisiología , Actinas/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Proteínas de Microfilamentos/metabolismo , Modelos Biológicos , Neuritas/efectos de los fármacos , Neuritas/enzimología , Neuronas/enzimología , Neuronas/metabolismo , Células PC12 , ARN Interferente Pequeño/fisiología , Ratas , Ratas Sprague-Dawley , Receptor trkA/metabolismo , Transducción de Señal , Transfección
13.
Cancer Res ; 67(18): 8596-604, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17875699

RESUMEN

The transcription factor NF-E2-related factor 2 (Nrf2) translocates into the nucleus and activates phase II genes encoding detoxification enzymes and antioxidant proteins, resulting in the protection of cells from oxidative insults. However, the involvement of Nrf2-mediated oxidative stress responses in breast cancer cells is largely unknown. Notably, during our study of the Nrf2 pathway in breast cancer cells, we observed that the nuclear matrix protein NRP/B was expressed and colocalized with Nrf2 in these cells, suggesting that NRP/B is involved in Nrf2-mediated oxidative stress responses. The expression level of NRP/B was variable in different breast cancer cells and breast cancer tissues, and was found to be localized in the nucleus. NRP/B expression was increased after exposure to the oxidative stress agent, hydrogen peroxide (H(2)O(2)), particularly in the highly aggressive MDA-MB-231 breast cancer cells. Association of NRP/B with Nrf2 in vitro and in vivo was observed in MDA-MB-231 breast cancer cells, and this association was up-regulated upon exposure to H(2)O(2), but not to sodium nitroprusside, SIN-1, and DETA-NO. NRP/B also enhanced Nrf2-mediated NAD(P)H:quinine oxidoreductase 1 promoter activity. Thus, this study reveals that NRP/B enhances oxidative stress responses in breast cancer cells via the Nrf2 pathway, identifying a novel role of nuclear matrix protein(s) in oxidative stress responses.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas de Microfilamentos/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Neuropéptidos/metabolismo , Proteínas Nucleares/metabolismo , Animales , Neoplasias de la Mama/genética , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/farmacología , Proteínas de Microfilamentos/biosíntesis , Proteínas de Microfilamentos/genética , Factor 2 Relacionado con NF-E2/biosíntesis , Factor 2 Relacionado con NF-E2/genética , Neuropéptidos/biosíntesis , Neuropéptidos/genética , Donantes de Óxido Nítrico/farmacología , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Estrés Oxidativo , Transfección , Regulación hacia Arriba
14.
Biochim Biophys Acta ; 1768(9): 2049-59, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17555706

RESUMEN

Cannabinoids are compounds that can modulate neuronal functions and immune responses via their activity at the CB(1) receptor. We used (2)H NMR order parameters and relaxation rate determination to delineate the behavior of magnetically aligned phospholipid bilayers in the presence of several structurally distinct cannabinoid ligands. THC (Delta(9)-Tetrahydrocannabinol) and WIN-55,212-2 were found to lower the phase transition temperature of the DMPC and to destabilize their acyl chains leading to a lower average S(CD) ( approximately 0.13), while methanandamide and CP-55,940 exhibited unusual properties within the lipid bilayer resulting in a greater average S(CD) ( approximately 0.14) at the top of the phospholipid upper chain. The CB(1) antagonist AM281 had average S(CD) values that were higher than the pure DMPC lipids, indicating a stabilization of the lipid bilayer. R(1Z) versus |S(CD)|(2) plots indicated that the membrane fluidity is increased in the presence of THC and WIN-55,212-2. The interaction of CP-55,940 with a variety of zwitterionic and charged membranes was also assessed. The unusual effect of CP-55,940 was present only in bicelles composed of DMPC. These studies strongly suggest that cannabinoid action on the membrane depends upon membrane composition as well as the structure of the cannabinoid ligands.


Asunto(s)
Cannabinoides/química , Cannabinoides/farmacología , Dimiristoilfosfatidilcolina/química , Membrana Dobles de Lípidos/química , Liposomas/química , Fluidez de la Membrana/efectos de los fármacos , Hidrógeno , Cinética , Espectroscopía de Resonancia Magnética , Conformación Molecular , Transición de Fase
15.
Int J Oncol ; 32(3): 619-23, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18292939

RESUMEN

Neuroblastoma is the second most common pediatric malignancy. The clinical course of this disease ranges from spontaneous regression and good survival to highly malignant therapy-resistant tumors. There is a continuous need for genetic and biologic markers for the diverse clinical phenotypes observed in neuroblastoma patients. One of the known markers in neuroblastoma is expression of the CXCR4 chemokine receptor. CXCR4 expression correlates with high-stage disease, and the autocrine stimulation of CXCR4 by its ligand (CXCL12) was shown to be necessary for the survival of some neuroblastoma cells in vitro. However, the mechanisms responsible for activation of the CXCL12-CXCR4 autocrine pathway in neuroblastoma remain uncertain. Our previous findings suggest that Csk homologous kinase (CHK) is a physiological inhibitor of CXCR4 expression. Since CHK is highly expressed in neurons, we evaluated changes in CHK expression in human neuroblastoma. CHK protein expression was below detectable levels based on Western blot analyses in 13 out of 16 human neuroblastoma cell lines and in 6 out of 16 primary neuroblastoma tissues. When CHK expression was restored in IMR32 neuroblastoma cells by retrovirus-mediated cDNA transfer, diminished CXCR4 mRNA and protein levels were observed, as assessed by RT-PCR and flow cytometry analyses, respectively. Furthermore, exogenous expression of CHK markedly suppressed the mRNA levels and secretion of the CXCL12 chemokine from IMR32 cells as well as inhibited the growth rate of these cells. Taken together, our data strongly suggest that CHK is capable of inhibiting the CXCL12-CXCR4 pathway in neuroblastoma.


Asunto(s)
Quimiocina CXCL12/metabolismo , Neuroblastoma/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/fisiología , Receptores CXCR4/metabolismo , Proliferación Celular , Quimiocina CXCL12/genética , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Humanos , Neuroblastoma/genética , Proteínas Proto-Oncogénicas pp60(c-src)/genética , ARN Mensajero/metabolismo , Receptores CXCR4/genética , Transducción de Señal/genética , Transfección , Células Tumorales Cultivadas
16.
Mol Biol Cell ; 16(2): 824-34, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15574879

RESUMEN

Human alveolar macrophages (AMs) phagocytose Pneumocystis (Pc) organisms predominantly through mannose receptors, although the molecular mechanism mediating this opsonin-independent process is not known. In this study, using AMs from healthy individuals, Pc phagocytosis was associated with focal F-actin polymerization and Cdc42, Rac1, and Rho activation in a time-dependent manner. Phagocytosis was primarily dependent on Cdc42 and RhoB activation (as determined by AM transfection with Cdc42 and RhoB dominant-negative alleles) and mediated predominantly through mannose receptors (as determined by siRNA gene silencing of AM mannose receptors). Pc also promoted PAK-1 phosphorylation, which was also dependent on RhoGTPase activation. HIV infection of AMs (as a model for reduced mannose receptor expression and function) was associated with impaired F-actin polymerization, reduced Cdc42 and Rho activation, and markedly reduced PAK-1 phosphorylation in response to Pc organisms. In healthy AMs, Pc phagocytosis was partially dependent on PAK activation, but dependent on the Rho effector molecule ROCK. These data provide a molecular mechanism for AM mannose receptor-mediated phagocytosis of unopsonized Pc organisms that appears distinct from opsonin-dependent phagocytic receptors. Reduced AM mannose receptor-mediated Cdc42 and Rho activation in the context of HIV infection may represent a mechanism that contributes to the pathogenesis of opportunistic pneumonia.


Asunto(s)
Proteínas Portadoras/metabolismo , Infecciones por VIH/inmunología , Lectinas Tipo C/metabolismo , Macrófagos Alveolares/inmunología , Lectinas de Unión a Manosa/metabolismo , Receptores de Superficie Celular/metabolismo , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rhoB/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Western Blotting , Líquido del Lavado Bronquioalveolar/citología , Broncoscopía , Proteínas Portadoras/genética , Células Cultivadas , Proteínas del Citoesqueleto , Activación Enzimática , Ensayo de Inmunoadsorción Enzimática , Fluoresceína-5-Isotiocianato , Colorantes Fluorescentes , Silenciador del Gen , Infecciones por VIH/complicaciones , Infecciones por VIH/virología , VIH-1/aislamiento & purificación , Humanos , Inmunidad Innata , Activación de Macrófagos , Macrófagos Alveolares/microbiología , Receptor de Manosa , Microscopía Confocal , Fagocitosis/efectos de los fármacos , Fagocitosis/fisiología , Pneumocystis/fisiología , Pruebas de Precipitina , Proteínas Serina-Treonina Quinasas/metabolismo , ARN Interferente Pequeño/metabolismo , Acetato de Tetradecanoilforbol/farmacología , Transfección , Zimosan/farmacología , Proteína de Unión al GTP cdc42/genética , Quinasas p21 Activadas , Proteínas de Unión al GTP rho , Proteína de Unión al GTP rhoB/genética
17.
Cancer Res ; 66(11): 5757-62, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16740714

RESUMEN

Amplification of the HER-2/neu (ErbB2) gene is observed in approximately 30% of human breast cancers, correlating with a poor clinical prognosis. Src kinases are also involved in the etiology of breast cancer, and their activation was suggested to be necessary for Neu-induced oncogenesis. To address whether Src activity is essential for Neu-mediated tumorigenesis, we used a physiologic inhibitor of Src kinase activity, the Csk homologous kinase (CHK), expressed as a mammary tissue-specific transgene. Our data, using a physiologic inhibitor of Src activity (CHK), showed that blocking of Neu-induced Src activity without altering Src expression levels had no significant effects on Neu-mediated mammary tumorigenesis in vivo. This contradicts the current paradigm that activation of Src kinases is essential for Neu-induced oncogenesis. This study is the first to distinguish between the kinase-dependent and kinase-independent actions of Src and shows that its kinase-dependent properties are not requisite for Neu-induced tumorigenesis.


Asunto(s)
Glándulas Mamarias Animales/enzimología , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Familia-src Quinasas/metabolismo , Animales , Femenino , Genes erbB-2/fisiología , Glándulas Mamarias Animales/crecimiento & desarrollo , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/genética , Virus del Tumor Mamario del Ratón/genética , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/genética , Fosforilación , Familia-src Quinasas/antagonistas & inhibidores
19.
PLoS Med ; 4(6): e186, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17550303

RESUMEN

BACKGROUND: While vascular endothelial growth factor (VEGF) expression in breast tumors has been correlated with a poor outcome in the pathogenesis of breast cancer, the expression, localization, and function of VEGF receptors VEGFR1 (also known as FLT1) and VEGFR2 (also known as KDR or FLK1), as well as neuropilin 1 (NRP1), in breast cancer are controversial. METHODS AND FINDINGS: We investigated the expression and function of VEGF and VEGF receptors in breast cancer cells. We observed that VEGFR1 expression was abundant, VEGFR2 expression was low, and NRP1 expression was variable. MDA-MB-231 and MCF-7 breast cancer cells, transfected with antisense VEGF cDNA or with siVEGF (VEGF-targeted small interfering RNA), showed a significant reduction in VEGF expression and increased apoptosis as compared to the control cells. Additionally, specifically targeted knockdown of VEGFR1 expression by siRNA (siVEGFR1) significantly decreased the survival of breast cancer cells through down-regulation of protein kinase B (AKT) phosphorylation, while targeted knockdown of VEGFR2 or NRP1 expression had no effect on the survival of these cancer cells. Since a VEGFR1-specific ligand, placenta growth factor (PGF), did not, as expected, inhibit the breast cancer cell apoptosis induced by siVEGF, and since VEGFR1 antibody also had no effects on the survival of these cells, we examined VEGFR1 localization. VEGFR1 was predominantly expressed internally in MDA-MB-231 and MCF-7 breast cancer cells. Specifically, VEGFR1 was found to be colocalized with lamin A/C and was expressed mainly in the nuclear envelope in breast cancer cell lines and primary breast cancer tumors. Breast cancer cells treated with siVEGFR1 showed significantly decreased VEGFR1 expression levels and a lack of VEGFR1 expression in the nuclear envelope. CONCLUSIONS: This study provides, to our knowledge for the first time, evidence of a unique survival system in breast cancer cells by which VEGF can act as an internal autocrine (intracrine) survival factor through its binding to VEGFR1. These results may lead to an improved strategy for tumor therapy based on the inhibition of angiogenesis.


Asunto(s)
Apoptosis/fisiología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Apoptosis/genética , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Citometría de Flujo , Humanos , Inmunohistoquímica , Inmunoprecipitación , Lamina Tipo A/metabolismo , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Desnudos , Mutación , Membrana Nuclear/metabolismo , Fosforilación , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/fisiología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 1 de Factores de Crecimiento Endotelial Vascular/fisiología
20.
Cell Signal ; 18(6): 871-81, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16168623

RESUMEN

Substantial evidence exists supporting the notion that Csk and CHK, two negative regulatory kinases of the Src tyrosine kinase family, play distinct roles during development of the nervous system. One of the differences relies on the effects of both kinases on the MAPK transduction pathway. Specifically, CHK was shown to enhance MAPK signaling, while the role of Csk was unclear. In this work, we compared the effect of CHK versus Csk on MAPK signaling and elucidated the signaling pathway mediated by CHK leading to the activation of Erk1/2. Exogenous expression of wild-type CHK, but not Csk or a dead-kinase mutant of CHK, resulted in enhanced Erk1/2 phosphorylation in PC12 cells. CHK inhibited Src activity following stimulation of the cells with NGF. However, stimulation of Erk1/2 activation by CHK was independent of the NGF stimulation or the inhibition of Src kinase by CHK. CHK induced a complex formation between SHP-2 and Grb2, subsequently leading to the increased activity of Ras as well as Erk1/2 activation via the Raf/MEK1/2 pathway. Down-regulation of the expression of endogenous CHK by RNAi in PC12 cells led to a significant decrease in MAPK activation following NGF stimulation. Stimulation of CHK-overexpressing PC12 cells with EGF induced neurite outgrowth in the majority of cells. Taken together, this study describes for the first time the Src-independent actions of CHK and provides novel insights into CHK function in neural cells.


Asunto(s)
Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Células PC12/efectos de los fármacos , Proteínas Quinasas/fisiología , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Proteínas ras/metabolismo , Familia-src Quinasas/metabolismo , Adenoviridae/genética , Animales , Células Cultivadas , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Vectores Genéticos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Factor de Crecimiento Nervioso/farmacología , Células PC12/citología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteína Quinasa C/metabolismo , Proteínas Quinasas/genética , Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas pp60(c-src)/genética , Ratas , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA