Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Biol Rep ; 51(1): 427, 2024 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-38498238

RESUMEN

BACKGROUND: Drug resistance is one of the most critical problems in gastric cancer therapy. This study was performed to investigate the valproic acid effects on the proliferation of sensitive and resistant cell lines of human gastric cancer, and to explore the mechanism of the agent on multi drug resistance and apoptosis genes. METHODS: The cytotoxicity effect of valproic acid on the EPG85.257 and EPG85.257RDB cells was assessed by the MTT assay, and the IC50 concentration was evaluated. Apoptosis, genotoxicity, and drug resistance pump activity were evaluated using comet assay, Real-time PCR, and flow cytometry, respectively. Cell proliferation was assayed using a scratch test. RESULTS: Dose-dependent toxicity was recorded after treatment of cells with valproic acid. Valproic acid represented a significant growth inhibition on EPG85.257 cells with IC50 values of 5.84 µM and 4.78 µM after 48 h and 72 h treatment, respectively. In contrast, the drug-resistant counterpart represented 8.7 µM and 7.02 µM IC50 values after the same treatment time. Valproic acid induced PTEN, Bcl2, P53, Bax, P21, and caspase3 expression in EPG85.257 cells, whereas p21, p53, PTEN, and ABCB1 were overexpressed in EPG5.257RDB. Valproic acid hindered cell migration in both cell lines (P < 0.01). Valproate genotoxicity was significantly higher in the parent cells than in their resistant EPG85.257RDB counterparts. Valproate led to a 62% reduction in the daunorubicin efflux of the MDR1 pump activity. CONCLUSIONS: Valproate can affect drug resistance in gastric cancer via a unique mechanism independent of MDR1 expression.


Asunto(s)
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Ácido Valproico/farmacología , Resistencia a Antineoplásicos/genética , Proteína p53 Supresora de Tumor , Resistencia a Múltiples Medicamentos/genética , Apoptosis , Línea Celular Tumoral , Proteínas Relacionadas con la Autofagia/metabolismo , Proteínas Relacionadas con la Autofagia/farmacología , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/farmacología , Proteínas de Transporte Vesicular/uso terapéutico
2.
J Adv Res ; 44: 185-199, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36725189

RESUMEN

INTRODUCTION: Microglia and macrophages participate in hematoma clearance after intracerebral hemorrhage (ICH), thereby facilitating tissue restoration and neurological recovery. Triggering receptor expressed on myeloid cells 2 (Trem2) has been indicated as a major pathology-induced immune signaling hub on the microglial/macrophage surface. Soluble Trem2 (sTrem2), the proteolytic form of Trem2, is abundant in the body fluid and is positively correlated with the pathological process. OBJECTIVES: In the present study, we aimed to investigate the potential role of sTrem2 in hematoma resolution after ICH and to elucidate its underlying mechanisms. METHODS: We explored the biological functions of sTrem2 in the murine ICH brain by stereotaxic injection of recombinant sTrem2 protein or by adeno-associated virus-mediated expression. Erythrocyte phagocytosis was assessed using flow cytometry and immunofluorescence. Western blotting was performed to evaluate protein expression. Changes in behavior, sTrem2-induced down-stream pathway, and microglia were examined. RESULTS: sTrem2 impedes hematoma resolution and impairs functional motor and sensory recovery. Interestingly, sTrem2 bypasses full-length Trem2, negatively regulating microglial/macrophage erythrophagocytosis, and promotes an inflammatory phenotype, which is associated with reduced retromer levels and impaired recycling of the pro-erythrophagocytic receptor CD36. Rescue of retromer Vps35 abolishes the phagocytosis-inhibiting effects and lysosome-dependent CD36 degradation caused by sTrem2. CONCLUSION: These findings indicate sTrem2 as a negative factor against microglia/macrophage-mediated hematoma and related neuronal damage clearance, provide insight into the mechanisms by which erythrophagocytosis is regulated and how it may be impaired after ICH, and suggest that the anti-proteolytic activity of Trem2 can be explored for ICH therapy.


Asunto(s)
Hemorragia Cerebral , Linfohistiocitosis Hemofagocítica , Animales , Ratones , Hemorragia Cerebral/complicaciones , Hemorragia Cerebral/metabolismo , Hemorragia Cerebral/patología , Fagocitosis/fisiología , Macrófagos/metabolismo , Microglía/metabolismo , Microglía/patología , Hematoma/complicaciones , Hematoma/metabolismo , Linfohistiocitosis Hemofagocítica/complicaciones , Linfohistiocitosis Hemofagocítica/metabolismo , Linfohistiocitosis Hemofagocítica/patología , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/farmacología , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/farmacología , Receptores Inmunológicos/metabolismo
3.
Leuk Res ; 123: 106967, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36270092

RESUMEN

BACKGROUND: Bortezomib resistance hampers the long-term survival of multiple myeloma (MM) patients. Our previous study has proved that downregulated lncRNA MEG3 is associated with the poor clinical outcome in MM. However, the effect of MEG3 on the sensitivity of bortezomib in MM and its possible molecular mechanism remains muddled. METHODS: In this study, CCK8 and flow cytometry techniques were used to assess cell viability in MEG3 overexpressed MM cells after bortezomib treatment. The expression of autophagy-related protein LC3 and p62 were distinguished by Western blot, and the mCherry-GFP-LC3 puncta reflecting autophagy level was observed under fluorescence microscope. RNA immunoprecipitation (RIP) technology was used to detect the binding relationship of MEG3 and ATG2B to PTBP1. RESULTS: Increased toxicity of bortezomib and diminished autophagy level were found in MEG3 overexpressed MM cells. Mechanistically, we discovered that RNA-binding protein PTBP1 could bind to MEG3 and ATG2B by RIP assay. Upregulation of MEG3 promoted PTBP1 expression and inhibited the expression level of ATG2B, suggesting that MEG3 recruited PTBP1 and then decayed ATG2B expression. CONCLUSION: In summary, our study illustrated that MEG3 increased bortezomib sensitivity by hindering autophagy through the PTBP1/ATG2B axis, providing a new therapeutic target for bortezomib-resistant MM patients.


Asunto(s)
Autofagia , Bortezomib , MicroARNs , Mieloma Múltiple , ARN Largo no Codificante , Humanos , Apoptosis , Proteínas Relacionadas con la Autofagia/genética , Proteínas Relacionadas con la Autofagia/metabolismo , Proteínas Relacionadas con la Autofagia/farmacología , Bortezomib/farmacología , Ribonucleoproteínas Nucleares Heterogéneas , MicroARNs/genética , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Proteína de Unión al Tracto de Polipirimidina/genética , Proteína de Unión al Tracto de Polipirimidina/farmacología , ARN Largo no Codificante/genética , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/farmacología
4.
Genes Genomics ; 44(7): 823-832, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35583792

RESUMEN

BACKGROUND: CHMP1A, a member of the ESCRT-III complex family, has been indicated as a brand-new inhibitor gene of tumors. Our previous research has revealed that CHMP1A plays a vital role in the development and progression of renal cell carcinoma (RCC). OBJECTIVE: To investigate the potential target pathway of the regulation of the tumor cell growth by CHMP1A. METHODS: The effect of CHMP1A on mTOR pathway was elucidated by western blotting. The effect of CHMP1A on the expression of p53 was evaluated, and A498 cell growth was assessed by colony formation and MTT assays. The expression of p53 was knocked down by shRNA-p53, and the effect of CHMP1A on mTOR after knockdown of p53 was evaluated. The effect of CHMP1A on apoptosis and its relationship with MDM2 pathway were detected by western blotting and FCM. Finally, the relationship between the regulation of p53 by CHMP1A and the PI3K/mTOR pathway was detected. RESULTS: This study showed that the mTOR pathway was suppressed significantly in CHMP1A-overexpressing A498 and 786-0 cells; moreover, the enhanced expression of p53 and the reduced proliferation were shown in CHMP1A-overexpressing A498 cells. Furthermore, CHMP1A was able to regulate the PI3K/PTEN/mTOR and MDM2/p53 pathways in order to suppress RCC. In addition, CHMP1A regulated Bax and Bcl-2 via MDM2/p53 to induce the apoptosis of tumor cells and upregulated the expression of p53 via the PI3K/mTOR pathway. CONCLUSIONS: The results convey that CHMP1A-related suppression of RCC is closely related to the PI3K/mTOR/p53 pathway.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Humanos , Neoplasias Renales/genética , Neoplasias Renales/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Serina-Treonina Quinasas TOR/farmacología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/farmacología
5.
Environ Toxicol ; 37(9): 2259-2268, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35616188

RESUMEN

Plenty of evidence has shown that endocytosis plays a key role in cancer progression; however, its effects in the progression of cancer stem cells (CSCs) are still fragmentary. In the present study, we firstly identified that mammalian Eps15 homology domain protein 1 (EHD1), an endocytic and metastasis-associated gene, was upregulated in the 3D non-adherent spheres derived from glioma cells compared to that in the corresponding parental cells. Further functional experiments revealed that EHD1 knockdown reduced the CSC-like traits of glioma cells, which were evident by the decrease of sphere-formation ability, ALDH1 activity, and CSC markers' expression. Additionally, EHD1 knockdown attenuated the tumor-initiating ability of glioma cells in vivo. Furthermore, it was shown that EHD1 bound to CD44, enhanced CD44 stability, and prevented its total ubiquitination. Indeed, overexpression of CD44 rescued the inhibitory effects of EHD1 knockdown on the CSC-like traits of glioma cells. Finally, through the online dataset analysis, we found that EHD1 indeed exhibited a higher level in glioma tissues relative to that in normal tissues, and a positive correlation with CSC markers' expression in glioma tissues. Notably, EHD1 expression was negatively correlated with the overall survival and relapse-free survival of glioma patients. Thus, this work indicates that EHD1 might be a potent target for glioma progression, especially through breaking the EHD1-CD44 interaction.


Asunto(s)
Glioma , Receptores de Hialuranos , Células Madre Neoplásicas , Proteínas de Transporte Vesicular , Familia de Aldehído Deshidrogenasa 1 , Línea Celular Tumoral , Glioma/genética , Glioma/metabolismo , Humanos , Receptores de Hialuranos/genética , Receptores de Hialuranos/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/farmacología
6.
Gut ; 70(12): 2261-2272, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33441378

RESUMEN

OBJECTIVE: As a canonical membrane tethering factor, the function of synbindin has been expanding and indicated in immune response. Here, we investigated the role of synbindin in the regulation of toll-like receptor 4 (TLR4) signalling and macrophage response to microbiota during colitis. DESIGN: Three distinct mouse models allowing global, myeloid-specific or intestinal epithelial cell-specific synbindin heterozygous deletion were constructed and applied to reveal the function of synbindin during dextran sodium sulfate (DSS) colitis. Effects of synbindin on TLR4 signalling and macrophage activation in response to bacterial lipopolysaccharide (LPS) or Fusobacterium nucleatum were evaluated. The colocalisation and interaction between synbindin and Rab7b were determined by immunofluorescence and coimmunoprecipitation. Synbindin expression in circulating monocytes and intestinal mucosal macrophages of patients with active IBD was detected. RESULTS: Global synbindin haploinsufficiency greatly exacerbated DSS-induced intestinal inflammation. The increased susceptibility to DSS was abolished by gut microbiota depletion, while phenocopied by specific synbindin heterozygous deletion in myeloid cells rather than intestinal epithelial cells. Profoundly aberrant proinflammatory gene signatures and excessive TLR4 signalling were observed in macrophages with synbindin interference in response to bacterial LPS or Fusobacterium nucleatum. Synbindin was significantly increased in intestinal mucosal macrophages and circulating monocytes from both mice with DSS colitis and patients with active IBD. Interleukin 23 and granulocyte-macrophage colony-stimulating factor were identified to induce synbindin expression. Mechanistic characterisation indicated that synbindin colocalised and directly interacted with Rab7b, which coordinated the endosomal degradation pathway of TLR4 for signalling termination. CONCLUSION: Synbindin was a key regulator of TLR4 signalling and restrained the proinflammatory macrophage activation against microbiota during colitis.


Asunto(s)
Colitis/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Proteínas del Tejido Nervioso/farmacología , Receptor Toll-Like 4/efectos de los fármacos , Proteínas de Transporte Vesicular/farmacología , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Transducción de Señal , Proteínas de Unión a GTP rab7/efectos de los fármacos
7.
Sci Rep ; 10(1): 17953, 2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33087848

RESUMEN

Proteins involved in the spaciotemporal regulation of GLUT4 trafficking represent potential therapeutic targets for the treatment of insulin resistance and type 2 diabetes. A key regulator of insulin- and exercise-stimulated glucose uptake and GLUT4 trafficking is TBC1D1. This study aimed to identify proteins that regulate GLUT4 trafficking and homeostasis via TBC1D1. Using an unbiased quantitative proteomics approach, we identified proteins that interact with TBC1D1 in C2C12 myotubes including VPS13A and VPS13C, the Rab binding proteins EHBP1L1 and MICAL1, and the calcium pump SERCA1. These proteins associate with TBC1D1 via its phosphotyrosine binding (PTB) domains and their interactions with TBC1D1 were unaffected by AMPK activation, distinguishing them from the AMPK regulated interaction between TBC1D1 and AMPKα1 complexes. Depletion of VPS13A or VPS13C caused a post-transcriptional increase in cellular GLUT4 protein and enhanced cell surface GLUT4 levels in response to AMPK activation. The phenomenon was specific to GLUT4 because other recycling proteins were unaffected. Our results provide further support for a role of the TBC1D1 PTB domains as a scaffold for a range of Rab regulators, and also the VPS13 family of proteins which have been previously linked to fasting glycaemic traits and insulin resistance in genome wide association studies.


Asunto(s)
Proteínas Activadoras de GTPasa/farmacología , Transportador de Glucosa de Tipo 4/metabolismo , Homeostasis/efectos de los fármacos , Homeostasis/genética , Fibras Musculares Esqueléticas/metabolismo , Proteínas/farmacología , Proteínas de Transporte Vesicular/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Células Cultivadas , Diabetes Mellitus Tipo 2 , Proteínas Activadoras de GTPasa/fisiología , Células HEK293 , Humanos , Resistencia a la Insulina , Masculino , Ratones Transgénicos , Proteínas/fisiología , Proteínas de Transporte Vesicular/fisiología
8.
J Cell Mol Med ; 21(10): 2503-2513, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28401647

RESUMEN

Hepatic ischaemia/reperfusion (HIR) induces severe damage on hepatocyte cell membrane, which leads to hepatocyte death and the subsequent HIR injury. In this study, we investigated the role and the mechanism of mitsugumin-53 (MG53), a novel cell membrane repair protein, in protecting the liver against HIR injury. Rats were subjected to sham operation or 70% warm HIR with or without recombined MG53 (rhMG53), caudal vein-injected 2 hrs before inducing HIR. In vitro, cultured hepatocyte AML12 cells were subjected to hypoxia/reoxygenation (H/R) in the presence of rhMG53 and/or dysferlin gene shRNAs or adenovirus transfection. HIR resulted in severe liver injury manifested as severe liver histological changes and increased AST and ALT release. Post-ischaemic hepatic oxidative stress was significantly enhanced demonstrated by elevated dihydroethidium level, increased 4-hydroxynonenal, enhanced 15-F2t-isoprostane and decreased SOD activity. rhMG53 administration attenuated post-HIR liver injury, decreased liver oxidative stress and further enhanced dysferlin protein expression and its colocalization with MG53. Similarly, H/R induced AML12 cell injury and oxidative stress, which were abolished by either rhMG53 or dysferlin overexpression but were exacerbated by dysferlin gene knockdown. Dysferlin overexpression further increased H/R-induced increased colocalization of MG53 and dysferlin. In conclusion, MG53 was anchored by dysferlin to reduce oxidative stress and cell death and attenuate HIR injury.


Asunto(s)
Apoptosis/efectos de los fármacos , Membrana Celular/metabolismo , Disferlina/metabolismo , Hepatocitos/efectos de los fármacos , Proteínas Musculares/farmacología , Daño por Reperfusión/metabolismo , Proteínas de Transporte Vesicular/farmacología , Animales , Línea Celular , Disferlina/genética , Hepatocitos/citología , Hepatocitos/metabolismo , Masculino , Ratones , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Estrés Oxidativo/efectos de los fármacos , Sustancias Protectoras/metabolismo , Sustancias Protectoras/farmacología , Interferencia de ARN , Ratas Sprague-Dawley , Proteínas Recombinantes/farmacología , Daño por Reperfusión/genética , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
9.
J Invest Dermatol ; 137(8): 1774-1783, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28454729

RESUMEN

In this study, we report that TIP39, a parathyroid hormone ligand family member that was recently identified to be expressed in the skin, can induce decorin expression and enhance wound repair. Topical treatment of mice with TIP39 accelerated wound repair, whereas TIP39-deficient mice had delayed repair that was associated with formation of abnormal collagen bundles. To study the potential mechanism responsible for the action of TIP39 in the dermis, fibroblasts were cultured in three-dimensional collagen gels, a process that results in enhanced decorin expression unless activated to differentiate to adipocytes, whereupon these cells reduce expression of several proteoglycans, including decorin. Small interfering RNA-mediated silencing of parathyroid hormone 2 receptor (PTH2R), the receptor for TIP39, suppressed the expression of extracellular matrix-related genes, including decorin, collagens, fibronectin, and matrix metalloproteases. Skin wounds in TIP39-/- mice had decreased decorin expression, and addition of TIP39 to cultured fibroblasts induced decorin and increased phosphorylation and nuclear translocation of CREB. Fibroblasts differentiated to adipocytes and treated with TIP39 also showed increased decorin and production of chondroitin sulfate. Furthermore, the skin of PTH2R-/- mice showed abnormal extracellular matrix structure, decreased decorin expression, and skin hardness. Thus, the TIP39-PTH2R system appears to be a previously unrecognized mechanism for regulation of extracellular matrix formation and wound repair.


Asunto(s)
Decorina/genética , Regulación de la Expresión Génica , Proteínas Nucleares/farmacología , ARN/genética , Receptor de Hormona Paratiroídea Tipo 2/genética , Proteínas de Transporte Vesicular/farmacología , Cicatrización de Heridas/fisiología , Heridas y Lesiones/genética , Animales , Diferenciación Celular , Células Cultivadas , Decorina/biosíntesis , Modelos Animales de Enfermedad , Femenino , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Factores de Empalme de ARN , Proteínas de Unión al ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Hormona Paratiroídea Tipo 2/biosíntesis , Transducción de Señal , Piel/lesiones , Piel/metabolismo , Piel/patología , Cicatrización de Heridas/efectos de los fármacos , Heridas y Lesiones/metabolismo , Heridas y Lesiones/patología
10.
Drug Discov Ther ; 9(4): 282-8, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25843701

RESUMEN

Some peptides that are highly conserved between insects and mammals have anti-tumor action. Screening for inhibitors of cell growth from animal fluids may provide useful clues to anti-tumor drugs. Inducers of autophagy also have anti-tumor activity. The current authors recently studied a protein found in silkworm hemolymph, Niemann-Pick disease type C2 (NPC2). This protein, which is highly conserved among eukaryotes, was found to have anti-proliferative action on a silkworm cell line. The current study found that the silkworm NPC2 protein also inhibits the growth of FM3A murine breast cancer cells. In FM3A cells, silkworm NPC2 increased phosphorylation of AMP-activated protein kinase and decreased phosphorylation of Akt and mammalian target of rapamycin, which are regulators of autophagy. This study also found that NPC2 increased the amount of microtubule-associated protein light chain 3 (LC3)-II, an autophagosome marker, in FM3A cells. Silkworm NPC2 also induced an increase in the number of LC3-dots, a marker of pre-autophagic endosomes, in FM3A cells. When silkworm NPC2 was used to inhibit FM3A cell growth, that inhibition was attenuated by chloroquine, which inhibits autophagic activity by preventing lysosomal acidification. Murine NPC2 also inhibited growth and induced autophagy in FM3A cells. These findings suggest that NPC2 is involved in the induction and/or maintenance of autophagy and may help to elucidate the mechanisms underlying other neurodegenerative disorders such as Niemann-Pick disease.


Asunto(s)
Autofagia/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Proteínas de Transporte Vesicular/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Hemolinfa/fisiología , Ratones
11.
PLoS One ; 7(7): e40341, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22815741

RESUMEN

Nearly 200 million people are infected by hepatitis C virus (HCV) worldwide. For replicating the HCV genome, the membrane-associated machinery needs to be formed by both HCV non-structural proteins (including NS5B) and human host factors such as VAPB. Recently, the 99-residue VAPC, a splicing variant of VAPB, was demonstrated to inhibit HCV replication via binding to NS5B, thus acting as an endogenous inhibitor of HCV infection. So far, the structure of VAPC remains unknown, and its interaction with NS5B has not been biophysically characterized. In this study, we conducted extensive CD and NMR investigations on VAPC which led to several striking findings: 1) although the N-terminal 70 residues are identical in VAPC and VAPB, they constitute the characteristic ß-barrel MSP fold in VAPB, while VAPC is entirely unstructured in solution, only with helical-like conformations weakly populated. 2) VAPC is indeed capable of binding to NS5B, with an average dissociation constant (Kd) of ∼20 µM. Intriguingly, VAPC remains dynamic even in the complex, suggesting that the VAPC-NS5B is a "fuzzy complex". 3) NMR mapping revealed that the major binding region for NS5B is located over the C-terminal half of VAPC, which is composed of three discrete clusters, of which only the first contains the region identical in VAPC and VAPB. The second region containing ∼12 residues appears to play a key role in binding since mutation of 4 residues within this region leads to almost complete loss of the binding activity. 4) A 14-residue mimetic, VAPC-14 containing the second region, only has a ∼3-fold reduction of the affinity. Our study not only provides critical insights into how a human factor mediates the formation of the HCV replication machinery, but also leads to design of VAPC-14 which may be further used to explore the function of VAPC and to develop anti-HCV molecules.


Asunto(s)
Hepacivirus/efectos de los fármacos , Hepacivirus/fisiología , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/metabolismo , Proteínas no Estructurales Virales/metabolismo , Secuencia de Aminoácidos , Humanos , Datos de Secuencia Molecular , Fragmentos de Péptidos/metabolismo , Unión Proteica , Estructura Secundaria de Proteína , Proteínas de Transporte Vesicular/farmacología , Proteínas no Estructurales Virales/química
12.
Arthritis Res Ther ; 14(1): R14, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22264405

RESUMEN

INTRODUCTION: Interleukin-34 (IL-34) is a recently defined cytokine, showing a functional overlap with macrophage colony stimulating factor (M-CSF). This study was undertaken to address the expression of IL-34 in rheumatoid arthritis (RA) patients and to investigate its regulation and pathogenic role in RA. METHODS: IL-34 levels were determined in the RA synovium, synovial fluid (SF) and fibroblast-like synovial cells (FLS) by immunohistochemistry, real-time PCR, enzyme-linked immunosorbent assay and immunoblotting. RA activity was assessed using Disease Activity Score 28 (DAS28) activity in the plasma collected at baseline and one year after treatment. Conditioned media (CM) were prepared from RA FLS culture with tumor necrosis factor alpha (TNFα) for 24 hours and used for functional assay. RESULTS: IL-34 was expressed in the synovium, SF, and FLS from RA patients. The production of IL-34 in FLS was up-regulated by TNFα in RA samples compared with osteoarthritis (OA) patients. Importantly, the preferential induction of IL-34 rather than M-CSF by TNFα in RAFLS was mediated by the transcription factor nuclear factor kappa B (NF-κB) and activation of c-Jun N-terminal kinase (JNK). IL-34 elevation in plasma from RA patients was decreased after the administration of disease-modifying anti-rheumatic drugs (DMARDs) in accordance with a decrease in DAS28. CM from RAFLS cultured with TNFα promoted chemotactic migration of human peripheral blood mononuclear cells (PBMCs) and subsequent osteoclast (OC) formation, effects that were attenuated by an anti-IL-34 antibody. CONCLUSIONS: These data provide novel information about the production of IL-34 in RA FLS and indicate that IL-34 is an additional osteoclastogenic factor regulated by TNFα in RA, suggesting a discrete role of IL-34 in inflammatory RA diseases.


Asunto(s)
Artritis Reumatoide/metabolismo , Fibroblastos/metabolismo , Membrana Sinovial/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Adulto , Anciano , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Activación Enzimática/efectos de los fármacos , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Humanos , Immunoblotting , Inmunohistoquímica , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Masculino , Persona de Mediana Edad , FN-kappa B/metabolismo , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Líquido Sinovial/metabolismo , Membrana Sinovial/efectos de los fármacos , Membrana Sinovial/patología , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/farmacología
13.
Eur J Immunol ; 41(2): 437-49, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21268013

RESUMEN

Serglycin (SG) is a proteoglycan expressed by hematopoietic cells and is constitutively secreted by multiple myeloma (MM) cells. SG participates in the regulation of various inflammatory events. We found that SG secreted by human MM cell lines inhibits both the classical and lectin pathways of complement, without influencing alternative pathway activity. The inhibitory effect of SG is due to direct interactions with C1q and mannose-binding lectin (MBL). C1q-binding is mediated through the glycosaminoglycan moieties of SG, whereas MBL binds additionally to SG protein core. Interactions between SG and C1q as well as MBL are diminished in the presence of chondroitin sulfate type E. In addition, we localized the SG-binding site to the collagen-like stalk of C1q. Interactions between SG and C1q as well as MBL are ionic in character and only the interaction with MBL was found to be partially dependent on the presence of calcium. We found the serum levels of SG to be elevated in patients with MM compared to healthy controls. Moreover, we found that SG expressed from myeloma plasma cells protects these cells from complement activation induced by treatment with anti-thymocyte immunoglobulins. This might protect myeloma cells during immunotherapy and promote survival of malignant cells.


Asunto(s)
Vía Clásica del Complemento/efectos de los fármacos , Lectina de Unión a Manosa de la Vía del Complemento/efectos de los fármacos , Glicosaminoglicanos/metabolismo , Mieloma Múltiple/inmunología , Proteoglicanos/farmacología , Proteínas de Transporte Vesicular/farmacología , Anciano , Animales , Anticuerpos Antineoplásicos/inmunología , Sitios de Unión/inmunología , Línea Celular Tumoral , Complemento C1q/metabolismo , Complemento C3b/metabolismo , Complemento C4b/metabolismo , Vía Alternativa del Complemento/efectos de los fármacos , Vía Clásica del Complemento/inmunología , Lectina de Unión a Manosa de la Vía del Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Femenino , Glicosaminoglicanos/farmacología , Hemólisis/efectos de los fármacos , Hemólisis/inmunología , Humanos , Masculino , Lectina de Unión a Manosa/metabolismo , Persona de Mediana Edad , Mieloma Múltiple/sangre , Unión Proteica/efectos de los fármacos , Unión Proteica/inmunología , Proteoglicanos/sangre , Proteoglicanos/inmunología , Proteoglicanos/metabolismo , Conejos , Ovinos , Proteínas de Transporte Vesicular/sangre , Proteínas de Transporte Vesicular/inmunología , Proteínas de Transporte Vesicular/metabolismo
14.
EMBO J ; 26(5): 1199-210, 2007 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-17318189

RESUMEN

Actin polymerization plays a critical role in clathrin-mediated endocytosis in many cell types, but how polymerization is regulated is not known. Hip1R may negatively regulate actin assembly during endocytosis because its depletion increases actin assembly at endocytic sites. Here, we show that the C-terminal proline-rich domain of Hip1R binds to the SH3 domain of cortactin, a protein that binds to dynamin, actin filaments and the Arp2/3 complex. We demonstrate that Hip1R deleted for the cortactin-binding site loses its ability to rescue fully the formation of abnormal actin structures at endocytic sites induced by Hip1R siRNA. To determine when this complex might function during endocytosis, we performed live cell imaging. The maximum in vivo recruitment of Hip1R, clathrin and cortactin to endocytic sites was coincident, and all three proteins disappeared together upon formation of a clathrin-coated vesicle. Finally, we showed that Hip1R inhibits actin assembly by forming a complex with cortactin that blocks actin filament barbed end elongation.


Asunto(s)
Actinas/metabolismo , Cortactina/metabolismo , Endocitosis/fisiología , Proteínas de Transporte Vesicular/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Sitios de Unión , Cortactina/genética , Cortactina/farmacología , Dinaminas/metabolismo , Endocitosis/efectos de los fármacos , Células HeLa , Humanos , Proteína Huntingtina , Proteínas de Microfilamentos , Modelos Biológicos , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Unión Proteica , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/farmacología
15.
Neuron ; 52(3): 485-96, 2006 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-17088214

RESUMEN

A longstanding hypothesis is that ion channels are present in the membranes of synaptic vesicles and might affect neurotransmitter release. Here we demonstrate that TRPM7, a member of the transient receptor potential (TRP) ion channel family, resides in the membrane of synaptic vesicles of sympathetic neurons, forms molecular complexes with the synaptic vesicle proteins synapsin I and synaptotagmin I, and directly interacts with synaptic vesicular snapin. In sympathetic neurons, changes in TRPM7 levels and channel activity alter acetylcholine release, as measured by EPSP amplitudes and decay times in postsynaptic neurons. TRPM7 affects EPSP quantal size, an intrinsic property of synaptic vesicle release. Targeted peptide interference of TRPM7's interaction with snapin affects the amplitudes and kinetics of postsynaptic EPSPs. Thus, vesicular TRPM7 channel activity is critical to neurotransmitter release in sympathetic neurons.


Asunto(s)
Acetilcolina/metabolismo , Neuronas/fisiología , Sinapsis/metabolismo , Transmisión Sináptica/fisiología , Vesículas Sinápticas/fisiología , Canales Catiónicos TRPM/fisiología , Animales , Animales Recién Nacidos , Western Blotting/métodos , Células Cultivadas , Cricetinae , Cricetulus , Estimulación Eléctrica/métodos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/fisiología , Potenciales Postsinápticos Excitadores/efectos de la radiación , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Microscopía Inmunoelectrónica/métodos , Mutagénesis/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/ultraestructura , Técnicas de Placa-Clamp/métodos , ARN Interferente Pequeño/farmacología , Ratas , Ratas Wistar , Ganglio Cervical Superior/citología , Sinapsis/clasificación , Vesículas Sinápticas/ultraestructura , Canales Catiónicos TRPM/química , Transfección/métodos , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/farmacología
16.
Exp Cell Res ; 305(1): 63-73, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15777788

RESUMEN

Cell migration occurs as a highly-regulated cycle of cell polarization, membrane extension at the leading edge, adhesion, contraction of the cell body, and release from the extracellular matrix at the trailing edge. In this study, we investigated the involvement of SNARE-mediated membrane trafficking in cell migration. Using a dominant-negative form of the enzyme N-ethylmaleimide-sensitive factor as a general inhibitor of SNARE-mediated membrane traffic and tetanus toxin as a specific inhibitor of VAMP3/cellubrevin, we conducted transwell migration assays and determined that serum-induced migration of CHO-K1 cells is dependant upon SNARE function. Both VAMP3-mediated and VAMP3-independent traffic were involved in regulating this cell migration. Inhibition of SNARE-mediated membrane traffic led to a decrease in the protrusion of lamellipodia at the leading edge of migrating cells. Additionally, the reduction in cell migration resulting from the inhibition of SNARE function was accompanied by perturbation of a Rab11-containing alpha(5)beta(1) integrin compartment and a decrease in cell surface alpha(5)beta(1) without alteration to total cellular integrin levels. Together, these observations suggest that inhibition of SNARE-mediated traffic interferes with the intracellular distribution of integrins and with the membrane remodeling that contributes to lamellipodial extension during cell migration.


Asunto(s)
Membrana Celular/fisiología , Movimiento Celular/fisiología , Proteínas de Transporte Vesicular/antagonistas & inhibidores , Sustitución de Aminoácidos , Animales , Células CHO , Adhesión Celular/fisiología , Movimiento Celular/efectos de los fármacos , Polaridad Celular , Cricetinae , Fibronectinas/farmacología , Integrinas/análisis , Mutagénesis Sitio-Dirigida , Proteínas Sensibles a N-Etilmaleimida , Proteínas Recombinantes/metabolismo , Proteínas SNARE , Transfección , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/farmacología , Proteínas de Transporte Vesicular/fisiología
17.
J Neurophysiol ; 93(1): 178-88, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15604462

RESUMEN

We show that activation of postsynaptic inositol 1,4,5-tris-phosphate receptors (IP(3)Rs) with the IP(3)R agonist adenophostin A (AdA) produces large increases in AMPA receptor (AMPAR) excitatory postsynaptic current (EPSC) amplitudes at hippocampal CA1 synapses. Co-perfusion of the Ca(2+) chelator bis-(o-aminophenoxy)-N,N,N',N'-tetraacetic acid strongly inhibited AdA-enhanced increases in EPSC amplitudes. We examined the role of AMPAR insertion/anchoring in basal synaptic transmission. Perfusion of an inhibitor of synaptotagmin-soluble n-ethylmaleimide-sensitive factor attachment protein (SNAP) receptor SNARE-mediated exocytosis depressed basal EPSC amplitudes, whereas a peptide that inhibits GluR2/3 interactions with postsynaptic density-95 (PDZ) domain proteins glutamate receptor interacting protein (GRIP)/protein interacting with C-kinase-1 (PICK1) enhanced basal synaptic transmission. These results suggest that constitutive trafficking and anchoring of AMPARs help maintain basal synaptic transmission. The regulation of postsynaptic AMPAR trafficking involves synaptotagmin-SNARE-mediated vesicle exocytosis and interactions between AMPARs and the PDZ domains in GRIP/PICK1. We show that inhibitors of synaptotagmin-SNARE-mediated exocytosis, or interactions between AMPARs and GRIP/PICK1, attenuated AdA-enhanced increases in EPSC amplitudes. These results suggest that IP(3)R-mediated Ca(2+) release can enhance AMPAR EPSC amplitudes through mechanisms that involve AMPAR-PDZ interactions and/or synaptotagmin-SNARE-mediated receptor trafficking.


Asunto(s)
Adenosina/análogos & derivados , Calcio/metabolismo , Ácido Egtácico/análogos & derivados , Hipocampo/citología , Neuronas/metabolismo , Receptores de Glutamato/metabolismo , Sinapsis/metabolismo , Adenosina/farmacología , Animales , Animales Recién Nacidos , Agonistas de los Canales de Calcio/farmacología , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/farmacología , Proteínas Portadoras/farmacología , Quelantes/farmacología , Complemento C2/farmacología , Complemento C2a , Interacciones Farmacológicas , Ácido Egtácico/farmacología , Estimulación Eléctrica , Hipocampo/fisiología , Técnicas In Vitro , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/farmacología , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/efectos de los fármacos , Proteínas del Tejido Nervioso/farmacología , Proteínas del Tejido Nervioso/fisiología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Técnicas de Placa-Clamp/métodos , Ratas , Ratas Sprague-Dawley , Receptores AMPA/química , Receptores AMPA/metabolismo , Proteínas SNARE , Sinapsis/efectos de los fármacos , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/fisiología , Sinaptotagminas , Proteínas de Transporte Vesicular/química , Proteínas de Transporte Vesicular/farmacología
18.
Curr Genet ; 47(1): 18-28, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15551135

RESUMEN

Eukaryotic cells invest a large proportion of their genome in maintaining telomere length homeostasis. Among the 173 non-essential yeast genes found to affect telomere length, a large proportion is involved in vacuolar traffic. When mutated, these vacuolar protein-sorting (VPS) genes lead to telomeres shorter than those observed in the wild type. Using genetic analysis, we characterized the pathway by which VPS15, VPS34, VPS22, VPS23 and VPS28 affect the telomeres. Our results indicate that these VPS genes affect telomere length through a single pathway and that this effect requires the activity of telomerase and the Ku heterodimer, but not the activity of Tel1p or Rif2p. We present models to explain the link between vacuolar traffic and telomere length homeostasis.


Asunto(s)
Telomerasa/farmacología , Telómero/ultraestructura , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/farmacología , Levaduras/genética , Homeostasis , Transducción de Señal
19.
Toxicol Appl Pharmacol ; 201(2): 120-36, 2004 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-15541752

RESUMEN

Evidence suggests that the neurological defects (gait abnormalities, foot splay, and skeletal muscle weakness) associated with acrylamide (ACR) intoxication are mediated by impaired neurotransmission at central and peripheral synapses. ACR can form adducts with nucleophilic residues on proteins and thereby alter corresponding structure and function. To evaluate protein adduction in nerve terminals as a possible mechanism of action, recombinant N-ethylmaleimide sensitive factor (NSF) was exposed in vitro to ACR (10 micromol) and mass spectrometry (MS) was used to identify adduct sites. MS analyses demonstrated that ACR formed adducts with sulfhydryl groups on cysteine residues (carbamoylethylcysteine, or CEC) of NSF. Ex vivo incubation of whole brain synaptosomes with ACR (0.001-1.0 M) produced concentration-dependent increases in CEC that were inversely correlated to reductions in neurotransmitter release that occurred over the same neurotoxicant concentration range. In synaptosomes isolated from rats intoxicated at a higher (50 mg/kg per day x 3, 5, 8, or 11 days) or a lower (21 mg/kg per day x 14, 21, or 28 day) ACR dose rate, CEC levels increased progressively up to a moderate level of neurotoxicity. To identify protein adducts, synaptosomal proteins labeled by ex vivo 14C-ACR exposure were separated by gel electrophoresis and probed by immunoblot analysis. Results showed that NSF and the SNARE protein, SNAP-25, were tentative ACR targets. Subsequent experiments indicated that ACR exposure increased synaptosomal levels of the 7S SNARE core complex, which is consistent with inhibition of NSF, SNAP-25 function, or both. These data suggest that adduction of cysteine residues on NSF and certain SNARE proteins might be causally involved in the nerve terminal dysfunction induced by ACR.


Asunto(s)
Acrilamidas/metabolismo , Encéfalo/metabolismo , Proteínas/metabolismo , Sinaptosomas/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Cromatografía Líquida de Alta Presión , Cisteína/metabolismo , Electroforesis en Gel de Poliacrilamida , Cromatografía de Gases y Espectrometría de Masas , Masculino , Espectrometría de Masas , Proteínas Sensibles a N-Etilmaleimida , Proteómica , Ratas , Ratas Sprague-Dawley , Proteínas SNARE , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Transporte Vesicular/farmacología
20.
Proc Natl Acad Sci U S A ; 100(15): 8736-41, 2003 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-12853568

RESUMEN

Measuring the biophysical properties of macromolecular complexes at work is a major challenge of modern biology. The protein complex composed of vesicle-associated membrane protein 2, synaptosomal-associated protein of 25 kDa, and syntaxin 1 [soluble N-ethyl-maleimide-sensitive factor attachment protein receptor (SNARE) complex] is essential for docking and fusion of neurotransmitter-filled synaptic vesicles with the presynaptic membrane. To better understand the fusion mechanisms, we reconstituted the synaptic SNARE complex in the imaging chamber of an atomic force microscope and measured the interaction forces between its components. Each protein was tested against the two others, taken either individually or as binary complexes. This approach allowed us to determine specific interaction forces and dissociation kinetics of the SNAREs and led us to propose a sequence of interactions. A theoretical model based on our measurements suggests that a minimum of four complexes is probably necessary for fusion to occur. We also showed that the regulatory protein neuronal Sec1 injected into the atomic force microscope chamber prevented the complex formation. Finally, we measured the effect of tetanus toxin protease on the SNARE complex and its activity by on-line registration during tetanus toxin injection. These experiments provide a basis for the functional study of protein microdomains and also suggest opportunities for sensitive screening of drugs that can modulate protein-protein interactions.


Asunto(s)
Fusión de Membrana/fisiología , Proteínas de la Membrana/fisiología , Vesículas Sinápticas/fisiología , Antígenos de Superficie/química , Antígenos de Superficie/fisiología , Fenómenos Biofísicos , Biofisica , Técnicas In Vitro , Cinética , Sustancias Macromoleculares , Proteínas de la Membrana/química , Microscopía de Fuerza Atómica , Proteínas Munc18 , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/farmacología , Proteínas del Tejido Nervioso/fisiología , Unión Proteica , Proteínas R-SNARE , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas SNARE , Proteína 25 Asociada a Sinaptosomas , Sintaxina 1 , Toxina Tetánica/farmacología , Proteínas de Transporte Vesicular/farmacología , Proteínas de Transporte Vesicular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...