Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Infect Immun ; 92(1): e0038323, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38018994

RESUMEN

The virulence-associated protein A (VapA) produced by virulent Rhodococcus equi allows it to replicate in macrophages and cause pneumonia in foals. It is unknown how VapA interacts with mammalian cell receptors, but intracellular replication of avirulent R. equi lacking vapA can be restored by supplementation with recombinant VapA (rVapA). Our objectives were to determine whether the absence of the surface receptors Toll-like receptor 2 (TLR2), complement receptor 3 (CR3), or Fc gamma receptor III (FcγRIII) impacts R. equi phagocytosis and intracellular replication in macrophages, and whether rVapA restoration of virulence in R. equi is dependent upon these receptors. Wild-type (WT) murine macrophages with TLR2, CR3, or FcγRIII blocked or knocked out (KO) were infected with virulent or avirulent R. equi, with or without rVapA supplementation. Quantitative bacterial culture and immunofluorescence imaging were performed. Phagocytosis of R. equi was not affected by blockade or KO of TLR2 or CR3. Intracellular replication of virulent R. equi was not affected by TLR2, CR3, or FcγRIII blockade or KO; however, avirulent R. equi replicated in TLR2-/- and CR3-/- macrophages but not in WT and FcγRIII-/-. rVapA supplementation did not affect avirulent R. equi phagocytosis but promoted intracellular replication in WT and all KO cells. By demonstrating that TLR2 and CR3 limit replication of avirulent but not virulent R. equi and that VapA-mediated virulence is independent of TLR2, CR3, or FcγRIII, our study provides novel insights into the role of these specific surface receptors in determining the entry and intracellular fate of R. equi.


Asunto(s)
Infecciones por Actinomycetales , Rhodococcus equi , Animales , Ratones , Infecciones por Actinomycetales/metabolismo , Infecciones por Actinomycetales/microbiología , Proteínas Bacterianas/genética , Caballos , Macrófagos/microbiología , Mamíferos , Fagocitosis , Rhodococcus equi/genética , Rhodococcus equi/patogenicidad , Receptor Toll-Like 2/genética , Factores de Virulencia , Interacciones Huésped-Patógeno
2.
Vet Microbiol ; 257: 109069, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33862330

RESUMEN

Rhodococcus equi is a soil saprophytic bacterium and intracellular pathogen that causes pneumonia in foals. Strains of R. equi that are virulent in foals contain a plasmid that encodes a virulence-associated protein A (VapA) necessary for replication in macrophages. Because other intracellular pathogens survive and replicate inside amoebae, we postulated that the VapA-bearing plasmid (pVAPA) confers a survival advantage for R. equi against environmental predators like amoebae. To test this hypothesis, we compared phagocytosis by and survival in Acanthamoeba castellanii of isogenic strains of pVAPA-positive and pVAPA-negative R. equi. Phagocytosis of the pVAPA-negative strain by A. castellanii was significantly (P < 0.0001) greater than the pVAPA-positive strain. Intracellular replication of the pVAPA-positive strain in A. castellanii was significantly (P < 0.0001) greater than the pVAPA-negative strain during both 48 h and 9 days. These results indicate that the presence of the VapA plasmid reduces uptake and aids replication of R. equi in A. castellanii.


Asunto(s)
Acanthamoeba castellanii/microbiología , Fagocitosis , Plásmidos/genética , Rhodococcus equi/genética , Rhodococcus equi/patogenicidad , Infecciones por Actinomycetales/microbiología , Animales , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Enfermedades de los Caballos/microbiología , Caballos , Microscopía Confocal , Rhodococcus equi/fisiología , Virulencia , Factores de Virulencia
3.
iScience ; 24(3): 102192, 2021 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-33718841

RESUMEN

Phagocytosis and autophagy play critical roles in immune defense. The human fungal pathogen Cryptococcus neoformans (Cn) subverts host autophagy-initiation complex (AIC)-related proteins, to promote its phagocytosis and intracellular parasitism of host cells. The mechanisms by which the pathogen engages host AIC-related proteins remain obscure. Here, we show that the recruitment of host AIC proteins to forming phagosomes is dependent upon the activity of CD44, a host cell surface receptor that engages fungal hyaluronic acid (HA). This interaction elevates intracellular Ca2+ concentrations and activates CaMKKß and its downstream target AMPKα, which results in activation of ULK1 and the recruitment of AIC components. Moreover, we demonstrate that HA-coated beads efficiently recruit AIC components to phagosomes and CD44 interacts with AIC components. Taken together, these findings show that fungal HA plays a critical role in directing the internalization and productive intracellular membrane trafficking of a fungal pathogen of global importance.

4.
Sci Rep ; 11(1): 371, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33432084

RESUMEN

Vaccines and therapeutics using in vitro transcribed mRNA hold enormous potential for human and veterinary medicine. Transfection agents are widely considered to be necessary to protect mRNA and enhance transfection, but they add expense and raise concerns regarding quality control and safety. We found that such complex mRNA delivery systems can be avoided when transfecting epithelial cells by aerosolizing the mRNA into micron-sized droplets. In an equine in vivo model, we demonstrated that the translation of mRNA into a functional protein did not depend on the addition of a polyethylenimine (PEI)-derived transfection agent. We were able to safely and effectively transfect the bronchial epithelium of foals using naked mRNA (i.e., mRNA formulated in a sodium citrate buffer without a delivery vehicle). Endoscopic examination of the bronchial tree and histology of mucosal biopsies indicated no gross or microscopic adverse effects of the transfection. Our data suggest that mRNA administered by an atomization device eliminates the need for chemical transfection agents, which can reduce the cost and the safety risks of delivering mRNA to the respiratory tract of animals and humans.


Asunto(s)
Caballos , Rociadores Nasales , ARN Mensajero/administración & dosificación , Mucosa Respiratoria , Animales , Animales Recién Nacidos , Células Cultivadas , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/efectos adversos , Portadores de Fármacos/farmacocinética , Sistemas de Liberación de Medicamentos/efectos adversos , Sistemas de Liberación de Medicamentos/métodos , Sistemas de Liberación de Medicamentos/veterinaria , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Nebulizadores y Vaporizadores/veterinaria , Polietileneimina/administración & dosificación , Polietileneimina/química , ARN Mensajero/efectos adversos , ARN Mensajero/farmacocinética , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Transcripción Genética , Transfección/métodos , Transfección/veterinaria , Vacunas de ADN/administración & dosificación , Vacunas de ADN/efectos adversos , Vacunas de ADN/farmacocinética
5.
Autophagy ; 17(2): 420-438, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-31983267

RESUMEN

Mitochondria operate as a central hub for many metabolic processes by sensing and responding to the cellular environment. Developmental cues from the environment have been implicated in selective autophagy, or mitophagy, of mitochondria during cell differentiation and tissue development. Mitophagy occurring in this context, termed programmed mitophagy, responds to cell state rather than mitochondrial damage and is often accompanied by a metabolic transition. However, little is known about the mechanisms that engage and execute mitophagy under physiological or developmental conditions. As the mammary gland undergoes post-natal development and lactation challenges mitochondrial homeostasis, we investigated the contribution of mitochondria to differentiation of mammary epithelial cells (MECs). Using lactogenic differentiation of the HC11 mouse MEC line, we demonstrated that HC11 cells transition to a highly energetic state during differentiation by engaging both oxidative phosphorylation and glycolysis. Interestingly, this transition was lost when autophagy was inhibited with bafilomycin A1 or knockdown of Atg7 (autophagy related 7). To evaluate the specific targeting of mitochondria, we traced mitochondrial oxidation and turnover in vitro with the fluorescent probe, pMitoTimer. Indeed, we found that differentiation engaged mitophagy. To further evaluate the requirement of mitophagy during differentiation, we knocked down the expression of Prkn/parkin in HC11 cells. We found that MEC differentiation was impaired in shPrkn cells, implying that PRKN is required for MEC differentiation. These studies suggest a novel regulation of MEC differentiation through programmed mitophagy and provide a foundation for future studies of development and disease associated with mitochondrial function in the mammary gland.Abbreviations: AA: antimycin A; ATG5: autophagy related 5; BAF: bafilomycin A1; BNIP3: BCL2 interacting protein 3; BNIP3L/NIX: BCL2 interacting protein 3 like; COX8A: cytochrome c oxidase subunit 8A; CQ: chloroquine; CSN2: casein beta; ECAR: extracellular acidification rate; FCCP: trifluoromethoxy carbonylcyanide phenylhydrazone; FUNDC1: FUN14 domain containing 1; HIF1A: hypoxia inducible factor 1 subunit alpha; L1: lactation day 1; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; MEC: mammary epithelial cell; mitoQ: mitoquinol; mROS: mitochondrial reactive oxygen species; OCR: oxygen consumption rate; P: priming; P16: pregnancy day 16; PARP1: poly(ADP-ribose) polymerase 1; PINK1: PTEN induced kinase 1; PPARGC1A: PPARG coactivator 1 alpha; PRKN: parkin RBR E3 ubiquitin protein ligase; shNT: short hairpin non-targeting control; SQSTM1: sequestosome 1; STAT3: signal transducer and activator of transcription 3; TEM: transmission electron microscopy; TFAM: transcription factor A, mitochondrial; U: undifferentiated.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia/fisiología , Diferenciación Celular/fisiología , Células Epiteliales/metabolismo , Animales , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Mitofagia/fisiología , Especies Reactivas de Oxígeno/metabolismo
6.
Breast Cancer Res ; 21(1): 125, 2019 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-31775907

RESUMEN

BACKGROUND: Mutations in genes associated with homologous recombination (HR) increase an individual's risk of developing triple-negative breast cancer (TNBC). Although known for their role in repairing dsDNA breaks, HR repair elements also stabilize and restart stalled replication forks. Essential to these functions are RAD51 and its paralogs, each of which has a unique role in preventing replication fork collapse and restart. However, progress toward understanding the regulation of these factors has been slow. With such a pivotal role in the maintenance of genomic integrity, furthering our understanding of this pathway through the discovery of new factors involved in HR is important. Recently, we showed that singleminded-2s (SIM2s) is stabilized in response to dsDNA breaks and is required for effective HR. METHODS: Initial analysis of the effect loss of SIM2s has on replication stress resolution was conducted using DNA combing assays in established breast cancer cell lines. Further analysis was conducted via immunostaining to determine the effect loss of SIM2s has on factor recruitment. In vivo confirmation was achieved through the use of a mammary epithelial cell conditional knockout mouse model before SIM2s' role in RAD51 recruitment was determined by immunoblotting. RESULTS: Here, we show loss of SIM2s decreases replication fork stability, leading to fork collapse in response to genotoxic stress. Furthermore, loss of SIM2s results in aberrant separation of sister chromatids during mitosis, which has been previously shown to result in chromosomal fragmentation and aneuploidy. Interestingly, loss of SIM2s was shown to result in failure of RAD51 to localize to sites of replication stress in both breast cancer cell lines and primary mammary epithelial cells. Finally, we observed SIM2 is stabilized in response to genotoxic stress and interacts with RAD51, which is necessary for RAD51-DNA binding. CONCLUSIONS: Together, these results show a role for SIM2s in the resolution of replication stress and further characterize the necessity of SIM2s for effective RAD51 loading in response to DNA damage or stress, ultimately promoting genomic integrity and thus preventing the accumulation of cancer-promoting mutations.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Replicación del ADN , Recombinasa Rad51/metabolismo , Estrés Fisiológico , Animales , Línea Celular Tumoral , Cromosomas/genética , Cromosomas/metabolismo , Daño del ADN , Reparación del ADN , Células Epiteliales/metabolismo , Inestabilidad Genómica , Histonas/metabolismo , Humanos , Ratones , Unión Proteica , Origen de Réplica
7.
Cancer Med ; 8(17): 7385-7398, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31638742

RESUMEN

BACKGROUND: Podosomes are membrane-bound adhesive structures formed by actin remodeling. They are capable of extracellular matrix (ECM) degradation, which is a prerequisite for cancer cell invasion and metastasis. The signaling mechanism of podosome formation is still unknown in cancer. We previously reported that Nck adaptors regulate directional cell migration and endothelial lumen formation by actin remodeling, while deficiency of Nck reduces cancer metastasis. This study evaluated the role of Nck adaptors in podosome biogenesis and cancer invasion. METHODS: This study was conducted in vitro using both healthy cells (Human Umbilical Vein Endothelial Cell, 3T3 fibroblasts) and cancer cells (prostate cancer cell line; PC3, breast cancer cell line; MDA-MB-231). Confocal and TIRF imaging of cells expressing Green Fluorescence Protein (GFP)  mutant under altered levels of Nck or downstream of kinase 1 (Dok1) was used to evaluate the podosome formation and fluorescent gelatin matrix degradation. Levels of Nck in human breast carcinoma tissue sections were detected by immune histochemistry using Nck polyclonal antibody. Biochemical interaction of Nck/Dok1 was detected in podosome forming cells using immune precipitation and far-western blotting. RESULTS: This study demonstrates that ectopic expression of Nck1 and Nck2 can induce the endothelial podosome formation in vitro. Nck silencing by short-hairpin RNA blocked podosome biogenesis and ECM degradation in cSrc-Y530F transformed endothelial cells in this study. Immunohistochemical analysis revealed the Nck overexpression in human breast carcinoma tissue sections. Immunoprecipitation and far-western blotting revealed the biochemical interaction of Nck/p62Dok in podosome forming cells. CONCLUSIONS: Nck adaptors in interaction with Dok1 induce podosome biogenesis and ECM degradation facilitating cancer cell invasion, and therefore a bona fide target of cancer therapy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Unión al ADN/metabolismo , Matriz Extracelular/patología , Neoplasias/patología , Proteínas Oncogénicas/metabolismo , Fosfoproteínas/metabolismo , Podosomas/metabolismo , Proteínas de Unión al ARN/metabolismo , Células 3T3 , Animales , Línea Celular Tumoral , Movimiento Celular , Matriz Extracelular/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ratones , Invasividad Neoplásica/patología
8.
Sci Rep ; 9(1): 14534, 2019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31601922

RESUMEN

Small intestinal damage induced by nonsteroidal anti-inflammatory drugs (NSAIDs) remains an under-recognized clinical disorder. The incomplete understanding of the pathophysiology has hampered the development of prevention and treatment strategies leading to the high morbidity and mortality rates. NSAIDs are known to modulate macroautophagy, a process indispensable for intestinal homeostasis. Whether NSAIDs stimulate or repress macroautophagy and how this correlates with the clinical manifestations of NSAID enteropathy, however, remains unknown. The objectives of this study were to determine whether NSAIDs impaired macroautophagy and how this affects macroautophagy-regulated intestinal epithelial cell (IEC) processes essential for intestinal homeostasis (i.e., clearance of invading pathogens, secretion and composition of mucus building blocks, and inflammatory response). We show that NSAID treatment of IECs inhibits macroautophagy in vitro and in vivo. This inhibition was likely attributed to a reduction in the area and/or distribution of lysosomes available for degradation of macroautophagy-targeted cargo. Importantly, IEC regulatory processes necessary for intestinal homeostasis and dependent on macroautophagy were dysfunctional in the presence of NSAIDs. Since macroautophagy is essential for gastrointestinal health, NSAID-induced inhibition of macroautophagy might contribute to the severity of intestinal injury by compromising the integrity of the mucosal barrier, preventing the clearance of invading microbes, and exacerbating the inflammatory response.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Células Epiteliales/citología , Intestinos/fisiopatología , Macroautofagia , Animales , Enfermedades Gastrointestinales/metabolismo , Enfermedades Gastrointestinales/microbiología , Células Caliciformes/metabolismo , Homeostasis , Indometacina/uso terapéutico , Inflamación , Interleucina-18/metabolismo , Intestinos/citología , Lisosomas/metabolismo , Ratones , Ratones Endogámicos C57BL , Infecciones por Salmonella/tratamiento farmacológico
9.
Oncogene ; 38(14): 2611-2626, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30531838

RESUMEN

There is increasing evidence that genomic instability is a prerequisite for cancer progression. Here we show that SIM2s, a member of the bHLH/PAS family of transcription factors, regulates DNA damage repair through enhancement of homologous recombination (HR), and prevents epithelial-mesenchymal transitions (EMT) in an Ataxia-telangiectasia mutated (ATM)-dependent manner. Mechanistically, we found that SIM2s interacts with ATM and is stabilized through ATM-dependent phosphorylation in response to IR. Once stabilized, SIM2s interacts with BRCA1 and supports RAD51 recruitment to the site of DNA damage. Loss of SIM2s through the introduction of shSIM2 or the mutation of SIM2s at one of the predicted ATM phosphorylation sites (S115) reduces HR efficiency through disruption of RAD51 recruitment, resulting in genomic instability and induction of EMT. The EMT induced by the mutation of S115 is characterized by a decrease in E-cadherin and an induction of the basal marker, K14, resulting in increased invasion and metastasis. Together, these results identify a novel player in the DNA damage repair pathway and provides a link in ductal carcinoma in situ progression to invasive ductal carcinoma through loss of SIM2s, increased genomic instability, EMT, and metastasis.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Transición Epitelial-Mesenquimal/genética , Recombinación Homóloga/genética , Animales , Proteína BRCA1/genética , Cadherinas/genética , Carcinoma Intraductal no Infiltrante/genética , Línea Celular Tumoral , Daño del ADN/genética , Reparación del ADN/genética , Femenino , Inestabilidad Genómica/genética , Humanos , Células MCF-7 , Ratones , Ratones Desnudos , Fosforilación/genética , Recombinasa Rad51/genética
10.
Inorg Chem ; 57(20): 12777-12786, 2018 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-30239197

RESUMEN

Ru(II)-polypyridyl complexes exhibit antitumor properties that can be systematically tailored by means of adjusting the ligand environment. In this work, the effect of incorporating π-extended moieties into anionic N∧O- based chelating ligands on the cytotoxic properties of Ru compounds is explored. Four new Ru(II) complexes, [Ru(bpy)2(dphol)][PF6] (1; bpy = 2,2'-bipyridine, dphol = dibenzo[ a, c]phenazin-10-olate), [Ru(phen)2(dphol)][PF6] (2; phen = 1,10-phenanthroline), [Ru(bpy)2(hbtz)][PF6] (3; hbtz = 2-(benzo[ d]thiazol-2-yl)phenolate), and [Ru(phen)2(hbtz)][PF6] (4) were synthesized and thoroughly characterized. In vitro cytotoxicity was investigated in human lung adenocarcinoma (A549) cells, which revealed that 4 is the most cytotoxic compound (IC50 = 0.8 µM) in the series including a control compound [Ru(bpy)2(quo)][PF6] (5; quo = 8-hydroxyquinolinate) and is nearly 8-fold more cytotoxic than cisplatin. An investigation of the mechanism of cell death led to the finding that compounds 1-4 disrupt the mitochondrial transmembrane potential (ΔΨm) in a concentration-dependent fashion, which is an event associated with the intrinsic pathway of apoptosis. Moreover, compound 4 triggers the activity of caspase-3/7, which eventually induces the apoptotic cellular death of A549 cells. Thus, increasing the overall lipophilicity of the Ru compounds by introducing π-extended moieties in the anionic N∧O- ligand is a successful strategy for realizing a new family of pro-apoptotic compounds with a [RuIIN5O]+ coordination environment.


Asunto(s)
Adenocarcinoma/metabolismo , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Redes y Vías Metabólicas/efectos de los fármacos , Nitrógeno/química , Compuestos de Rutenio/farmacología , Células A549 , Supervivencia Celular/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Humanos , Ligandos , Neoplasias Pulmonares/metabolismo , Modelos Moleculares , Estructura Molecular , Nitrógeno/metabolismo , Compuestos de Rutenio/química
11.
Cancer Res ; 78(14): 3899-3912, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29769200

RESUMEN

Ras signaling originates from transient nanoscale compartmentalized regions of the plasma membrane composed of specific proteins and lipids. The highly specific lipid composition of these nanodomains, termed nanoclusters, facilitates effector recruitment and therefore influences signal transduction. This suggests that Ras nanocluster proteolipid composition could represent a novel target for future chemoprevention interventions. There is evidence that consumption of fish oil containing long-chain n-3 polyunsaturated fatty acids (n-3 PUFA) such as eicosapentaenoic acid (EPA, 20:5Δ5,8,11,14,17) and docosahexaenoic acid (DHA, 22:6Δ4,7,10,13,16,19) may reduce colon cancer risk in humans, yet the mechanism underlying this effect is unknown. Here, we demonstrate that dietary n-3 PUFA reduce the lateral segregation of cholesterol-dependent and -independent nanoclusters, suppressing phosphatidic acid-dependent oncogenic KRas effector interactions, via their physical incorporation into plasma membrane phospholipids. This results in attenuation of oncogenic Ras-driven colonic hyperproliferation in both Drosophila and murine models. These findings demonstrate the unique properties of dietary n-3 PUFA in the shaping of Ras nanoscale proteolipid complexes and support the emerging role of plasma membrane-targeted therapies.Significance: The influence of dietary long chain n-3 polyunsaturated fatty acids on plasma membrane protein nanoscale organization and KRas signaling supports development of plasma membrane-targeted therapies in colon cancer.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/14/3899/F1.large.jpg Cancer Res; 78(14); 3899-912. ©2018 AACR.


Asunto(s)
Membrana Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ácidos Grasos Omega-3/farmacología , Proteolípidos/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Animales , Membrana Celular/metabolismo , Células Cultivadas , Colesterol/metabolismo , Dieta , Ácidos Docosahexaenoicos/farmacología , Drosophila/metabolismo , Ácido Eicosapentaenoico/análogos & derivados , Ácido Eicosapentaenoico/farmacología , Aceites de Pescado , Ratones , Fosfolípidos/metabolismo
12.
Br J Nutr ; 119(2): 163-175, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29249211

RESUMEN

Cell membrane fatty acids influence fundamental properties of the plasma membrane, including membrane fluidity, protein functionality, and lipid raft signalling. Evidence suggests that dietary n-3 PUFA may target the plasma membrane of immune cells by altering plasma membrane lipid dynamics, thereby regulating the attenuation of immune cell activation and suppression of inflammation. As lipid-based immunotherapy might be a promising new clinical strategy for the treatment of inflammatory disorders, we conducted in vitro and in vivo experiments to examine the effects of n-3 PUFA on CD4+ T cell membrane order, mitochondrial bioenergetics and lymphoproliferation. n-3 PUFA were incorporated into human primary CD4+ T cells phospholipids in vitro in a dose-dependent manner, resulting in a reduction in whole cell membrane order, oxidative phosphorylation and proliferation. At higher doses, n-3 PUFA induced unique phase separation in T cell-derived giant plasma membrane vesicles. Similarly, in a short-term human pilot study, supplementation of fish oil (4 g n-3 PUFA/d) for 6 weeks in healthy subjects significantly elevated EPA (20 : 5n-3) levels in CD4+ T cell membrane phospholipids, and reduced membrane lipid order. These results demonstrate that the dynamic reshaping of human CD4+ T cell plasma membrane organisation by n-3 PUFA may modulate down-stream clonal expansion.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/ultraestructura , Membrana Celular/efectos de los fármacos , Ácidos Grasos Omega-3/farmacología , Anciano , Anciano de 80 o más Años , Membrana Celular/química , Membrana Celular/fisiología , Grasas de la Dieta/administración & dosificación , Suplementos Dietéticos , Ácido Eicosapentaenoico/sangre , Metabolismo Energético/efectos de los fármacos , Ácidos Grasos/sangre , Femenino , Aceites de Pescado/administración & dosificación , Humanos , Activación de Linfocitos/efectos de los fármacos , Masculino , Lípidos de la Membrana/sangre , Lípidos de la Membrana/química , Persona de Mediana Edad , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Fosfolípidos/sangre , Fosfolípidos/química , Proyectos Piloto
13.
Mol Biol Cell ; 28(24): 3500-3516, 2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-28954862

RESUMEN

Although it is known that noncatalytic region of tyrosine kinase (Nck) regulates cell adhesion and migration by bridging tyrosine phosphorylation with cytoskeletal remodeling, the role of Nck in tumorigenesis and metastasis has remained undetermined. Here we report that Nck is required for the growth and vascularization of primary tumors and lung metastases in a breast cancer xenograft model as well as extravasation following injection of carcinoma cells into the tail vein. We provide evidence that Nck directs the polarization of cell-matrix interactions for efficient migration in three-dimensional microenvironments. We show that Nck advances breast carcinoma cell invasion by regulating actin dynamics at invadopodia and enhancing focalized extracellular matrix proteolysis by directing the delivery and accumulation of MMP14 at the cell surface. We find that Nck-dependent cytoskeletal changes are mechanistically linked to enhanced RhoA but restricted spatiotemporal activation of Cdc42. Using a combination of protein silencing and forced expression of wild-type/constitutively active variants, we provide evidence that Nck is an upstream regulator of RhoA-dependent, MMP14-mediated breast carcinoma cell invasion. By identifying Nck as an important driver of breast carcinoma progression and metastasis, these results lay the groundwork for future studies assessing the therapeutic potential of targeting Nck in aggressive cancers.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Neoplasias de la Mama/metabolismo , Proteínas Oncogénicas/deficiencia , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Transformación Celular Neoplásica , Femenino , Xenoinjertos , Humanos , Metaloproteinasa 14 de la Matriz/metabolismo , Ratones , Metástasis de la Neoplasia , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Fosforilación , Podosomas/metabolismo , Transducción de Señal , Proteína de Unión al GTP rhoA/metabolismo
14.
Mol Pharmacol ; 91(3): 178-188, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28007926

RESUMEN

The circadian clock plays a role in many biologic processes, yet very little is known about its role in metabolism of drugs and carcinogens. The purpose of this study was to define the impact of circadian rhythms on benzo-a-pyrene (BaP) metabolism in the mouse mammary gland and develop a circadian in vitro model for investigating changes in BaP metabolism resulting from cross-talk between the molecular clock and aryl hydrocarbon receptor. Female 129sv mice (12 weeks old) received a single gavage dose of 50 mg/kg BaP at either noon or midnight, and mammary tissues were isolated 4 or 24 hours later. BaP-induced Cyp1a1 and Cyp1b1 mRNA levels were higher 4 hours after dosing at noon than at 4 hours after dosing at midnight, and this corresponded with parallel changes in Per gene expression. In our in vitro model, we dosed MCF10A mammary cells at different times after serum shock to study how time of day shifts drug metabolism in cells. Analysis of CYP1A1 and CYP1B1 gene expression showed the maximum enzyme-induced metabolism response 12 and 20 hours after shock, as determined by ethoxyresorufin-O-deethylase activity, metabolism of BaP, and formation of DNA-BaP adducts. The pattern of PER-, BMAL-, and aryl hydrocarbon receptor-induced P450 gene expression and BaP metabolism was similar to BaP-induced Cyp1A1 and Cyp1B1 and molecular clock gene expression in mouse mammary glands. These studies indicate time-of-day exposure influences BaP metabolism in mouse mammary glands and describe an in vitro model that can be used to investigate the circadian influence on the metabolism of carcinogens.


Asunto(s)
Benzo(a)pireno/metabolismo , Mama/citología , Ritmo Circadiano , Aductos de ADN/metabolismo , Glándulas Mamarias Animales/citología , Animales , Biomarcadores/metabolismo , Mama/metabolismo , Proteínas CLOCK/genética , Proteínas CLOCK/metabolismo , Línea Celular , Ritmo Circadiano/genética , Citocromo P-450 CYP1A1/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Ratones , Modelos Biológicos
15.
Biochim Biophys Acta ; 1858(1): 85-96, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26476105

RESUMEN

The mechanisms by which n-3 polyunsaturated fatty acids (n-3 PUFA), abundant in fish oil, exert their anti-inflammatory effects have not been rigorously defined. We have previously demonstrated that n-3 PUFA decrease the amount of phosphatidylinositol-(4,5)-bisphosphate, [PI(4,5)P2], in CD4(+) T cells, leading to suppressed actin remodeling upon activation. Since discrete pools of PI(4,5)P2 exist in the plasma membrane, we determined whether n-3 PUFA modulate spatial organization of PI(4,5)P2 relative to raft and non-raft domains. We used Förster resonance energy transfer (FRET) to demonstrate that lipid raft mesodomains in the plasma membrane of CD4(+) T cells enriched in n-3 PUFA display increased co-clustering of Lck(N10) and LAT(ΔCP), markers of lipid rafts. CD4(+) T cells enriched in n-3 PUFA also exhibited a depleted plasma membrane non-raft PI(4,5)P2 pool as detected by decreased co-clustering of Src(N15), a non-raft marker, and PH(PLC-δ), a PI(4,5)P2 reporter. Incubation with exogenous PI(4,5)P2 rescued the effects on the non-raft PI(4,5)P2 pool, and reversed the suppression of T cell proliferation in CD4(+) T cells enriched with n-3 PUFA. Furthermore, CD4(+) T cells isolated from mice fed a 4% docosahexaenoic acid (DHA)-enriched diet exhibited a decrease in the non-raft pool of PI(4,5)P2, and exogenous PI(4,5)P2 reversed the suppression of T cell proliferation. Finally, these effects were not due to changes to post-translational lipidation, since n-3 PUFA did not alter the palmitoylation status of signaling proteins. These data demonstrate that n-3 PUFA suppress T cell proliferation by altering plasma membrane topography and the spatial organization of PI(4,5)P2.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Grasas de la Dieta/farmacología , Ácidos Docosahexaenoicos/farmacología , Microdominios de Membrana/efectos de los fármacos , Fosfatidilinositol 4,5-Difosfato/química , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Biomarcadores/metabolismo , Linfocitos T CD4-Positivos/química , Linfocitos T CD4-Positivos/citología , Expresión Génica , Vectores Genéticos , Lentivirus/genética , Microdominios de Membrana/química , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilinositol 4,5-Difosfato/farmacología , Fosfolipasa C delta/genética , Fosfolipasa C delta/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Cultivo Primario de Células , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
16.
Mol Biol Cell ; 26(17): 3047-60, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26157164

RESUMEN

Multiple angiogenic cues modulate phosphotyrosine signaling to promote vasculogenesis and angiogenesis. Despite its functional and clinical importance, how vascular cells integrate phosphotyrosine-dependent signaling to elicit cytoskeletal changes required for endothelial morphogenesis remains poorly understood. The family of Nck adaptors couples phosphotyrosine signals with actin dynamics and therefore is well positioned to orchestrate cellular processes required in vascular formation and remodeling. Culture of endothelial cells in three-dimensional collagen matrices in the presence of VEGF stimulation was combined with molecular genetics, optical imaging, and biochemistry to show that Nck-dependent actin remodeling promotes endothelial cell elongation and proper organization of VE-cadherin intercellular junctions. Major morphogenetic defects caused by abrogation of Nck signaling included loss of endothelial apical-basal polarity and impaired lumenization. Time-lapse imaging using a Förster resonance energy transfer biosensor, immunostaining with phospho-specific antibodies, and GST pull-down assays showed that Nck determines spatiotemporal patterns of Cdc42/aPKC activation during endothelial morphogenesis. Our results demonstrate that Nck acts as an important hub integrating angiogenic cues with cytoskeletal changes that enable endothelial apical-basal polarization and lumen formation. These findings point to Nck as an emergent target for effective antiangiogenic therapy.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Proteínas Oncogénicas/metabolismo , Citoesqueleto de Actina/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Línea Celular , Células Endoteliales de la Vena Umbilical Humana , Humanos , Uniones Intercelulares/metabolismo , Proteínas Oncogénicas/genética , Fosfotirosina/metabolismo , Transducción de Señal , Análisis Espacio-Temporal , Imagen de Lapso de Tiempo
17.
Biol Reprod ; 92(3): 67, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25568306

RESUMEN

Environmental exposure to endocrine-disrupting chemicals (EDCs) is one cause of premature ovarian failure (POF). Hexavalent chromium (CrVI) is a heavy metal EDC widely used in more than 50 industries, including chrome plating, welding, wood processing, and tanneries. Recent data from U.S. Environmental Protection Agency indicate increased levels of Cr in drinking water from several American cities, which potentially predispose residents to various health problems. Recently, we demonstrated that gestational exposure to CrVI caused POF in F1 offspring. The current study was performed to identify the molecular mechanism behind CrVI-induced POF. Pregnant rats were treated with 25 ppm of potassium dichromate from Gestational Day (GD) 9.5 to GD 14.5 through drinking water, and the fetuses were exposed to CrVI through transplacental transfer. Ovaries were removed from the fetuses or pups on Embryonic Day (ED) 15.5, ED 17.5, Postnatal Day (PND) 1, PND 4, or PND 25, and various analyses were performed. Results showed that gestational exposure to CrVI: 1) increased germ cell/oocyte apoptosis and advanced germ cell nest (GCN) breakdown; 2) increased X-prolyl aminopeptidase (Xpnpep) 2, a POF marker in humans, during GCN breakdown; 3) decreased Xpnpep2 during postnatal follicle development; and 4) increased colocalization of Xpnpep2 with Col3 and Col4. We also found that Xpnpep2 inversely regulated the expression of Col1, Col3, and Col4 in all the developmental stages studied. Thus, CrVI advanced GCN breakdown and increased follicle atresia in F1 female progeny by targeting Xpnpep2.


Asunto(s)
Aminopeptidasas/fisiología , Cromo/efectos adversos , Cromo/farmacología , Fase Folicular/efectos de los fármacos , Óvulo/efectos de los fármacos , Insuficiencia Ovárica Primaria/inducido químicamente , Insuficiencia Ovárica Primaria/fisiopatología , Animales , Apoptosis/efectos de los fármacos , Carcinógenos Ambientales/efectos adversos , Carcinógenos Ambientales/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Colágeno Tipo I/fisiología , Colágeno Tipo III/fisiología , Colágeno Tipo IV/fisiología , Modelos Animales de Enfermedad , Femenino , Atresia Folicular/efectos de los fármacos , Atresia Folicular/fisiología , Fase Folicular/fisiología , Ovario/efectos de los fármacos , Ovario/fisiología , Óvulo/fisiología , Embarazo , Ratas
18.
J Nutr ; 144(8): 1306-13, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24944284

RESUMEN

Cluster of differentiation 4(+) (CD4(+)) effector T-cell subsets [e.g., T-helper (Th) 1 and Th17] are implicated in autoimmune and inflammatory disorders such as multiple sclerosis, psoriasis, and rheumatoid arthritis. Interleukin (IL)-6 is a pleiotropic cytokine that induces Th17 polarization via signaling through the membrane-bound transducer glycoprotein 130 (GP130). Previously, we demonstrated that n-3 (ω-3) polyunsaturated fatty acids (PUFAs) reduce CD4(+) T-cell activation and differentiation into pathogenic Th17 cells by 25-30%. Here we report that n-3 PUFAs alter the response of CD4(+) T cells to IL-6 in a lipid raft membrane-dependent manner. Naive splenic CD4(+) T cells from fat-1 transgenic mice exhibited 30% lower surface expression of the IL-6 receptor. This membrane-bound receptor is known to be shed during cellular activation, but the release of soluble IL-6 receptor after treatment with anti-CD3 and anti-CD28 was not changed in the CD4(+) T cells from fat-1 mice, suggesting that the decrease in surface expression was not due to ectodomain release. We observed a significant 20% decrease in the association of GP130 with lipid rafts in activated fat-1 CD4(+) T cells and a 35% reduction in GP130 homodimerization, an obligate requirement for downstream signaling. The phosphorylation of signal transducer and activator of transcription 3 (STAT3), a downstream target of IL-6-dependent signaling, was also decreased by 30% in response to exogenous IL-6 in fat-1 CD4(+) T cells. Our results suggest that n-3 PUFAs suppress Th17 cell differentiation in part by reducing membrane raft-dependent responsiveness to IL-6, an essential polarizing cytokine.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Ácidos Grasos Omega-3/farmacología , Interleucina-6/metabolismo , Células Th17/efectos de los fármacos , Animales , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Receptor gp130 de Citocinas/genética , Receptor gp130 de Citocinas/metabolismo , Femenino , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Fosforilación , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Interleucina-6/genética , Receptores de Interleucina-6/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Bazo/efectos de los fármacos , Bazo/metabolismo
19.
J Am Chem Soc ; 136(22): 7861-4, 2014 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-24854400

RESUMEN

The new dirhodium compound [Rh2(µ-O2CCH3)2(η(1)-O2CCH3)(phenbodipy)(H2O)3][O2CCH3] (1), which incorporates a bodipy fluorescent tag, was prepared and studied by confocal fluorescence microscopy in human lung adenocarcinoma (A549) cells. It was determined that 1 localizes mainly in lysosomes and mitochondria with no apparent nuclear localization in the 1-100 µM range. These results support the conclusion that cellular organelles rather than the nucleus can be targeted by modification of the ligands bound to the Rh2(4+) core. This is the first study of a fluorophore-labeled metal-metal bonded compound, work that opens up new venues for the study of intracellular distribution of dinuclear transition metal anticancer complexes.


Asunto(s)
Adenocarcinoma/metabolismo , Colorantes Fluorescentes/química , Neoplasias Pulmonares/metabolismo , Metales/química , Metales/metabolismo , Rodio/química , Compuestos de Boro , Línea Celular Tumoral , Humanos , Ligandos , Lisosomas/química , Lisosomas/metabolismo , Microscopía Confocal , Microscopía Fluorescente , Mitocondrias/química , Mitocondrias/metabolismo
20.
PLoS One ; 9(3): e90908, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24651647

RESUMEN

Dietary supplementation with natural chemoprotective agents is receiving considerable attention because of health benefits and lack of toxicity. In recent in vivo and in vitro experimental studies, diets rich in n-3 polyunsaturated fatty acids have been shown to provide significant anti-tumor action. In this investigation, the effects of control fatty acids (oleic acid (OA), linoleic acid (LA)) and n-3 PUFA, e.g., docosahexaenoic acid (DHA) on the uptake and metabolism of the carcinogenic polycyclic aromatic hydrocarbon, benzo[a]pyrene (BaP) was investigated in A549 cells, a human adenocarcinoma alveolar basal epithelial cell line. A549 cells activate BaP through the cytochrome P450 enzyme system to form reactive metabolites, a few of which covalently bind to DNA and proteins. Therefore, multiphoton microscopy spectral analysis combined with linear unmixing was used to identify the parent compound and BaP metabolites formed in cells, in the presence and absence of fatty acids. The relative abundance of select metabolites was associated with altered P450 activity as determined using ethoxyresorufin-O-deethylase activity in cells cultured in the presence of BSA-conjugated fatty acids. In addition, the parent compound within cellular membranes increases significantly in the presence of each of the fatty acids, with the greatest accumulation observed following DHA treatment. DHA treated cells exhibit significantly lower pyrene-like metabolites indicative of lower adducts including DNA adducts compared to control BSA, OA or LA treated cells. Further, DHA reduced the abundance of the proximate carcinogen BaP 7,8-dihydrodiol and the 3-hydroxybenzo[a]pyrene metabolites compared to other treatments. The significant changes in BaP metabolites in DHA treated cells may be mediated by the effects on the physicochemical properties of the membrane known to affect enzyme activity related to phase I and phase II metabolism. In summary, DHA is a highly bioactive chemo-protective agent capable of modulating BaP-induced DNA adducts.


Asunto(s)
Adenocarcinoma/metabolismo , Benzo(a)pireno/metabolismo , Ácidos Grasos/farmacología , Neoplasias Pulmonares/metabolismo , Adenocarcinoma del Pulmón , Animales , Butionina Sulfoximina/farmacología , Bovinos , Línea Celular Tumoral , Citocromo P-450 CYP1A1/metabolismo , Aductos de ADN/metabolismo , Ácidos Docosahexaenoicos/farmacología , Glutatión/metabolismo , Humanos , Fotones , Albúmina Sérica Bovina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...