Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cornea ; 43(6): 784-789, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38437155

RESUMEN

PURPOSE: The purpose of this study was to highlight characteristic clinical and microscopic findings and report the long-term follow-up of pediatric excimer laser-assisted penetrating keratoplasty (excimer-PKP) for congenital stromal corneal dystrophy (CSCD). METHODS: A 2-year-old Greek child presented with CSCD at our department. Clinical examination showed bilateral flake-like whitish corneal opacities affecting the entire corneal stroma up to the limbus. Genetic testing identified a mutation of the decorin gene (c.962delA). The variant was not present in the parents and represented a de novo mutation. The uncorrected visual acuity was 20/100 in both eyes. Excimer-PKP (8.0/8.1 mm) was performed on the right eye at the age of 2.5 years and on the left eye at the age of 3 years. Postoperatively, alternating occlusion treatment was performed. RESULTS: The light microscopic examination demonstrated a disorganized extracellular matrix of the corneal stroma characterized by a prominent irregular arrangement of stromal collagen lamellae with large interlamellar clefts containing ground substance, highlighted by periodic acid-Schiff- and Alcian blue-positive reaction detecting acid mucopolysaccharides. Electron microscopy showed disorganization and caliber variation of collagen lamellae and thin filaments within an electron-lucent ground substance. The postoperative course was unremarkable. Both grafts remained completely clear 14 years postoperatively. Corneal tomography showed moderate regular astigmatism with normal corneal thickness. The corrected distance visual acuity was 20/25 in both eyes. CONCLUSIONS: Excimer-PKP for CSCD might be associated with excellent long-term results and a good prognosis, particularly when the primary surgery is performed at a very young age. However, this requires close postoperative follow-up examinations by an experienced pediatric ophthalmologist to avoid severe amblyopia.


Asunto(s)
Distrofias Hereditarias de la Córnea , Queratoplastia Penetrante , Láseres de Excímeros , Agudeza Visual , Humanos , Queratoplastia Penetrante/métodos , Estudios de Seguimiento , Distrofias Hereditarias de la Córnea/cirugía , Distrofias Hereditarias de la Córnea/fisiopatología , Preescolar , Agudeza Visual/fisiología , Láseres de Excímeros/uso terapéutico , Masculino , Sustancia Propia/cirugía , Sustancia Propia/patología , Femenino , Decorina/genética
2.
Nat Commun ; 15(1): 1277, 2024 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-38341434

RESUMEN

Overexpression of the transmembrane matrix metalloproteinase MT1-MMP/MMP14 promotes cancer cell invasion. Here we show that MT1-MMP-positive cancer cells turn MT1-MMP-negative cells invasive by transferring a soluble catalytic ectodomain of MT1-MMP. Surprisingly, this effect depends on the presence of TKS4 and TKS5 in the donor cell, adaptor proteins previously implicated in invadopodia formation. In endosomes of the donor cell, TKS4/5 promote ADAM-mediated cleavage of MT1-MMP by bridging the two proteases, and cleavage is stimulated by the low intraluminal pH of endosomes. The bridging depends on the PX domains of TKS4/5, which coincidently interact with the cytosolic tail of MT1-MMP and endosomal phosphatidylinositol 3-phosphate. MT1-MMP recruits TKS4/5 into multivesicular endosomes for their subsequent co-secretion in extracellular vesicles, together with the enzymatically active ectodomain. The shed ectodomain converts non-invasive recipient cells into an invasive phenotype. Thus, TKS4/5 promote intercellular transfer of cancer cell invasiveness by facilitating ADAM-mediated shedding of MT1-MMP in acidic endosomes.


Asunto(s)
Metaloproteinasa 14 de la Matriz , Neoplasias , Humanos , Metaloproteinasa 14 de la Matriz/genética , Metaloproteinasa 14 de la Matriz/metabolismo , Péptido Hidrolasas/metabolismo , Neoplasias/genética , Endosomas/metabolismo , Invasividad Neoplásica , Línea Celular Tumoral
3.
BMC Cancer ; 23(1): 650, 2023 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-37438741

RESUMEN

BACKGROUND: Recent reports have demonstrated that the entire mitochondrial genome can be secreted in extracellular vesicles (EVs), but the biological attributes of this cell-free mitochondrial DNA (mtDNA) remain insufficiently understood. We used next-generation sequencing to compare plasma EV-derived mtDNA to that of whole blood (WB), peripheral blood mononuclear cells (PBMCs), and formalin-fixed paraffin-embedded (FFPE) tumor tissue from eight rectal cancer patients and WB and fresh-frozen (FF) tumor tissue from eight colon cancer patients. METHODS: Total DNA was isolated before the mtDNA was enriched by PCR with either two primer sets generating two long products or multiple primer sets (for the FFPE tumors), prior to the sequencing. mtDNA diversity was assessed as the total variant number, level of heteroplasmy (mutant mtDNA copies mixed with wild-type copies), variant distribution within the protein-coding genes, and the predicted functional effect of the variants in the different sample types. Differences between groups were compared by paired Student's t-test or ANOVA with Dunnett's multiple comparison tests when comparing matched samples from patients. Mann-Whitney U test was used when comparing differences between the cancer types and patient groups. Pearson correlation analysis was performed. RESULTS: In both cancer types, EV mtDNA presented twice as many variants and had significantly more low-level heteroplasmy than WB mtDNA. The EV mtDNA variants were clustered in the coding regions, and the proportion of EV mtDNA variants that were missense mutations (i.e., estimated to moderately affect the mitochondrial protein function) was significantly higher than in WB and tumor tissues. Nonsense mutations (i.e., estimated to highly affect the mitochondrial protein function) were only observed in the tumor tissues and EVs. CONCLUSION: Taken together, plasma EV mtDNA in CRC patients exhibits a high degree of diversity. TRIAL REGISTRATION: ClinicalTrials.gov: NCT01816607 . Registered 22 March 2013.


Asunto(s)
Ácidos Nucleicos Libres de Células , Neoplasias del Colon , Vesículas Extracelulares , Genoma Mitocondrial , Humanos , Leucocitos Mononucleares , Secuenciación de Nucleótidos de Alto Rendimiento , ADN Mitocondrial/genética , Ácidos Nucleicos Libres de Células/genética , Vesículas Extracelulares/genética , Proteínas Mitocondriales
4.
Cell Mol Life Sci ; 80(7): 177, 2023 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-37285022

RESUMEN

Cells release extracellular vesicles (EVs) of different sizes. Small EVs (< 200 nm) can originate from the fusion of multivesicular bodies with the plasma membrane, i.e. exosomes, and from budding of the plasma membrane, i.e. small ectosomes. To investigate the molecular machinery required for the release of small EVs, we developed a sensitive assay based on incorporation of radioactive cholesterol in EV membranes and used it in a siRNA screening. The screening showed that depletion of several SNARE proteins affected the release of small EVs. We focused on SNAP29, VAMP8, syntaxin 2, syntaxin 3 and syntaxin 18, the depletion of which reduced the release of small EVs. Importantly, this result was verified using gold standard techniques. SNAP29 depletion resulted in the largest effect and was further investigated. Immunoblotting analysis of small EVs showed that the release of several proteins considered to be associated with exosomes like syntenin, CD63 and Tsg101 was reduced, while the level of several proteins that have been shown to be released in ectosomes (annexins) or by secretory autophagy (LC3B and p62) was not affected by SNAP29 depletion. Moreover, these proteins appeared in different fractions when the EV samples were further separated by a density gradient. These results suggest that SNAP29 depletion mainly affects the secretion of exosomes. To investigate how SNAP29 affects exosome release, we used microscopy to study the distribution of MBVs using CD63 labelling and CD63-pHluorin to detect fusion events of MVBs with the plasma membrane. SNAP29 depletion caused a redistribution of CD63-labelled compartments but did not change the number of fusion events. Further experiments are therefore needed to fully understand the function of SNAP29. To conclude, we have developed a novel screening assay that has allowed us to identify several SNAREs involved in the release of small EVs.


Asunto(s)
Exosomas , Vesículas Extracelulares , Exosomas/genética , Exosomas/metabolismo , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Cuerpos Multivesiculares/metabolismo , Autofagia
5.
J Cell Mol Med ; 24(17): 10258-10262, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32672902

RESUMEN

The receptor tyrosine kinase HER2 is associated with a number of human malignancies and is an important therapeutic target. The antibody-drug conjugate trastuzumab emtansine (T-DM1; Kadcyla® ) is recommended as a first-line treatment for patients with HER2-positive metastatic breast cancer. T-DM1 combines the antibody-induced effects of the anti-HER2 antibody trastuzumab (Herceptin® ) with the cytotoxic effect of the tubulin inhibitor mertansine (DM1). For DM1 to have effect, the T-DM1-HER2 complex has to be internalized and the trastuzumab part of T-DM1 has to be degraded. HER2 is, however, considered endocytosis-resistant. As a result of this, trastuzumab is only internalized to a highly limited extent, and if internalized, it is rapidly recycled. The exact reasons for the endocytosis resistance of HER2 are not clear, but it is stabilized by heat-shock protein 90 (Hsp90) and Hsp90 inhibitors induce internalization and degradation of HER2. HER2 can also be internalized upon activation of protein kinase C, and contrary to trastuzumab alone, the combination of two or more anti-HER2 antibodies can induce efficient internalization and degradation of HER2. With intention to find ways to improve the action of T-DM1, we investigated how different ways of inducing HER2 internalization leads to degradation of trastuzumab. The results show that although both Hsp90 inhibition and activation of protein kinase C induce internalization of trastuzumab, only Hsp90 inhibition induces degradation. Furthermore, we find that antibody internalization and degradation are increased when trastuzumab is combined with the clinically approved anti-HER2 antibody pertuzumab (Perjeta® ).


Asunto(s)
Ado-Trastuzumab Emtansina/farmacología , Anticuerpos Monoclonales Humanizados/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Trastuzumab/farmacología , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Maitansina/farmacología , Receptor ErbB-2/metabolismo
6.
Exp Cell Res ; 382(2): 111473, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31233741

RESUMEN

ErbB3, which belongs to the epidermal growth factor receptor (EGFR) or ErbB family of receptor tyrosine kinases, is involved in progression of several human cancers and a tight regulation of its expression is crucial. An important mechanism for regulation of ErbB proteins is endocytosis and we recently showed that ErbB3, contrary to other ErbB proteins, like EGFR and ErbB2, is constitutively internalized and degraded. Several studies show that protein kinase C (PKC) can regulate the activation, localization and stability of EGFR and ErbB2. Activation of PKC causes their down-regulation from the plasma membrane, but instead of being degraded the receptors accumulate in an endosomal recycling compartment. Since little is known about possible connections between ErbB3 and PKC, we have in the present study investigated effects PKC activity has on ErbB3 stability and intracellular trafficking. While PKC inhibition tends to increase ErbB3 degradation, activation of PKC causes ErbB3 stabilization. The stabilization was not due to inhibited internalization, on the contrary we find that expression of ErbB3 at the plasma membrane is reduced upon PMA-induced PKC activation. However, while endocytosed ErbB3 under normal conditions and upon PKC inhibition is found in early endosomal antigen 1 (EEA1) positive early endosomes and lysosomal-associated membrane protein 1 (LAMP1) positive late endosomes/lysosomes, indicating that it follows the classic degradative pathway, ErbB3 localizes to EEA1 and LAMP1 negative compartments upon PMA-induced activation of PKC. Altogether this shows that PKC regulates the stability of ErbB3, and knockdown experiments show that PKCδ is essential in this process. A likely explanation is that PKC regulates endosomal sorting of ErbB3 and that activated PKC sorts ErbB3 away from the degradative pathway.


Asunto(s)
Proteína Quinasa C-delta/metabolismo , Receptor ErbB-3/metabolismo , Carbazoles/farmacología , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Endocitosis/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Células MCF-7 , Neurregulina-1/farmacología , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Estabilidad Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteolisis/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Ubiquitinación/efectos de los fármacos
7.
Exp Cell Res ; 371(1): 139-150, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30098331

RESUMEN

Overexpression of ErbB2 is frequent in cancer and understanding the mechanisms which regulate its expression is important. ErbB2 is considered endocytosis resistant. It has no identified ligand, but upon heterodimerization it is a potent mediator of proliferative signaling. A recent study established a role for protein kinase C (PKC) in internalization and recycling of ErbB2. We have now further investigated the molecular mechanisms involved in PKC-mediated downregulation of ErbB2. We confirm that PMA-induced PKC activation causes ErbB2 internalization, but while the Hsp90 inhibitor 17-AAG induced ErbB2 degradation, PMA had no such effect. When combined with 17-AAG, PMA had additive effect on ErbB2 internalization indicating that Hsp90 inhibition and PKC activation induce internalization by alternative mechanisms. We confirm that while 17-AAG-induced internalization was clathrin-mediated, PMA-induced internalization was clathrin independent. This difference may be explained by while both 17-AAG and PMA reduced the constitutive tyrosine phosphorylation of ErbB2, only 17-AAG induced Hsp90 dissociation, Hsp70 recruitment and ubiquitination of ErbB2. Importantly, since PMA induced internalization of ErbB2, but not dissociation of Hsp90, Hsp90 does not per se retain ErbB2 at the plasma membrane. The morphology of the compartment into which receptors are sorted upon PKC activation has not previously been identified. By immuno-electron microscopy, we show that PMA sorts ErbB2 into a complex tubulovesicular or cisternal organelle resembling a previously described endocytic recycling compartment.


Asunto(s)
Benzoquinonas/farmacología , Regulación Neoplásica de la Expresión Génica , Proteínas HSP90 de Choque Térmico/genética , Lactamas Macrocíclicas/farmacología , Proteína Quinasa C/genética , Receptor ErbB-2/genética , Acetato de Tetradecanoilforbol/farmacología , Línea Celular Tumoral , Clatrina/genética , Clatrina/metabolismo , Endocitosis/efectos de los fármacos , Endocitosis/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Membranas Intracelulares/efectos de los fármacos , Membranas Intracelulares/metabolismo , Membranas Intracelulares/ultraestructura , Microscopía Inmunoelectrónica , Fosforilación/efectos de los fármacos , Proteína Quinasa C/metabolismo , Estabilidad Proteica/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteolisis/efectos de los fármacos , Receptor ErbB-2/metabolismo , Transducción de Señal , Ubiquitinación/efectos de los fármacos
8.
Biochim Biophys Acta Mol Cell Res ; 1864(12): 2241-2252, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28867611

RESUMEN

The ErbB family of receptor tyrosine kinases mediates activation of a wide network of signaling pathways. ErbB3 has weak kinase activity, but its six docking sites for the p85 subunit of phosphoinositide 3-kinase make it an important contributor to proliferative signaling. ErbB3 has a relatively short half-life but the exact mechanisms controlling its turnover are unclear as contradictory reports exist. ErbB-mediated signaling is, however, negatively regulated by endocytosis of the receptors, followed by either recycling or degradation. Our previous studies showed that ErbB3 can be endocytosed and degraded in the absence of its ligand heregulin. However, binding of heregulin increased the degradation rate. In the current study we have investigated in more detail the trafficking and degradation of ErbB3 in the presence or absence of heregulin. We report that ErbB3 is internalized by clathrin-mediated endocytosis both in the presence and absence of heregulin. Moreover, we show that both proteasomal and lysosomal activity regulate ErbB3 degradation. Although steady-state expression of ErbB3 is regulated by proteasomal activity to a large extent, probably linked to a previously identified ER-localized quantity control, the results indicate that internalization, both constitutive and ligand-induced, causes lysosomal degradation of ErbB3. Furthermore, we show that ErbB3 interacts with the ESCRT-0 subunit Hrs both in the presence and absence of heregulin. This indicates an ESCRT-mediated sorting of ErbB3 to late endosomes and lysosomes, and in line with this we show that impaired ESCRT function leads to an endosomal accumulation of ErbB3.


Asunto(s)
Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Neurregulina-1/metabolismo , Fosfoproteínas/metabolismo , Proteolisis , Receptor ErbB-3/metabolismo , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Endocitosis/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Humanos , Ligandos , Lisosomas/metabolismo , Microscopía Confocal , Neurregulina-1/genética , Fosfoproteínas/genética , Receptor ErbB-3/genética , Transducción de Señal
9.
J Cell Mol Med ; 20(10): 1999-2011, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27469139

RESUMEN

The human epidermal growth factor receptor 2 (HER2/ErbB2) is overexpressed in a number of human cancers. HER2 is the preferred heterodimerization partner for other epidermal growth factor receptor (EGFR) family members and is considered to be resistant to endocytic down-regulation, properties which both contribute to the high oncogenic potential of HER2. Antibodies targeting members of the EGFR family are powerful tools in cancer treatment and can function by blocking ligand binding, preventing receptor dimerization, inhibiting receptor activation and/or inducing receptor internalization and degradation. With respect to antibody-induced endocytosis of HER2, various results are reported, and the effect seems to depend on the HER2 expression level and whether antibodies are given as individual antibodies or as mixtures of two or more. In this study, the effect of a mixture of two monoclonal antibodies against non-overlapping epitopes of HER2 was investigated with respect to localization and stability of HER2. Individual antibodies had limited effect, but the combination of antibodies induced internalization and degradation of HER2 by multiple endocytic pathways. In addition, HER2 was phosphorylated and ubiquitinated upon incubation with the antibody combination, and the HER2 kinase activity was found to be instrumental in antibody-induced HER2 down-regulation.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Endocitosis , Epítopos/inmunología , Receptor ErbB-2/inmunología , Actinas/metabolismo , Anticuerpos Monoclonales/farmacología , Línea Celular , Clatrina/metabolismo , Dinaminas/metabolismo , Endocitosis/efectos de los fármacos , Humanos , Fosforilación/efectos de los fármacos , Pinocitosis/efectos de los fármacos , Polimerizacion/efectos de los fármacos , Proteolisis/efectos de los fármacos , Ubiquitinación/efectos de los fármacos
10.
Biochim Biophys Acta ; 1863(6 Pt A): 1179-88, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26975582

RESUMEN

BACKGROUND: In contrast to other members of the EGF receptor family, ErbB3 is constitutively internalized in a clathrin-dependent manner. Previous studies have shown that ErbB3 does not interact with the coated pit localized adaptor complex 2 (AP-2), and that ErbB3 lacks two AP-2 interacting internalization signals identified in the EGF receptor. Several other clathrin-associated sorting proteins which may recruit cargo into coated pits have, however, been identified, and the study was performed to identify adaptors needed for constitutive internalization of ErbB3. METHODS: A high-throughput siRNA screen was used to identify adaptor proteins needed for internalization of ErbB3. Upon knock-down of candidate proteins internalization of ErbB3 was identified using an antibody-based internalization assay combined with automatic fluorescence microscopy. RESULTS: Among 29 candidates only knock-down of epsin 1 turned out to inhibit ErbB3. Epsin 1 has ubiquitin interacting motifs (UIMs) and we show that ErbB3 interacts with an epsin 1 deletion mutant containing these UIMs. In support of an ErbB3-epsin 1 UIM dependent interaction, we show that ErbB3 is constitutively ubiquitinated, but that both ubiquitination and the ErbB3-epsin 1 interaction increase upon ligand binding. CONCLUSION: Altogether the results are consistent with a model whereby both constitutive and ligand-induced internalization of ErbB3 are regulated through interaction with epsin 1. GENERAL SIGNIFICANCE: Internalization is an important regulator of growth factor receptor mediated signaling and the current study identify mechanisms regulating plasma membrane turnover of ErbB3.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Clatrina/metabolismo , Endocitosis , Receptor ErbB-3/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Clatrina/genética , Células HeLa , Humanos , Immunoblotting , Células MCF-7 , Microscopía Confocal , Unión Proteica , Interferencia de ARN , Receptor ErbB-3/genética , Ubiquitinación
11.
PLoS One ; 11(2): e0149000, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26872261

RESUMEN

BACKGROUND AND PURPOSE: Previous studies of ischemia-reperfusion injury (IRI) in hearts from mice with cardiac-restricted overexpression of CCN2 have shown that CCN2 increases tolerance towards IRI. The objectives of this study were to investigate to what extent post-ischemic administration of recombinant human CCN2 (rhCCN2) would limit infarct size and improve functional recovery and what signaling pathways are involved. EXPERIMENTAL APPROACH: Isolated mice hearts were perfused ad modum Langendorff, subjected to no-flow, global ischemia, and subsequently, exposed to mammalian cell derived, full-length (38-40kDa) rhCCN2 (250 nM) or vehicle during the first 15 min of a 60 min reperfusion period. KEY RESULTS: Post-ischemic administration of rhCCN2 resulted in attenuation of infarct size from 58 ± 4% to 34 ± 2% (p < 0.001) which was abrogated by concomitant administration of the PI3 kinase inhibitor LY294002 (45 ± 3% vs. 50 ± 3%, ns). In congruence with reduction of infarct size rhCCN2 also improved recovery of left ventricular developed pressure (p < 0.05). Western blot analyses of extracts of ex vivo-perfused murine hearts also revealed that rhCCN2 evoked concentration-dependent increase of cardiac phospho-GSK3ß (serine-9) contents. CONCLUSIONS AND IMPLICATIONS: We demonstrate that post-ischemic administration of rhCCN2 increases the tolerance of ex vivo-perfused murine hearts to IRI. Mechanistically, this postconditioning effect of rhCCN2 appeared to be mediated by activation of the reperfusion injury salvage kinase pathway as demonstrated by sensitivity to PI3 kinase inhibition and increased CCN2-induced phosphorylation of GSK3ß (Ser-9). Thus, the rationale for testing rhCCN2-mediated post-ischemic conditioning of the heart in more complex models is established.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/uso terapéutico , Corazón/efectos de los fármacos , Poscondicionamiento Isquémico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Miocardio/patología , Animales , Células Cultivadas , Cromonas/uso terapéutico , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Poscondicionamiento Isquémico/métodos , Masculino , Ratones Endogámicos C57BL , Morfolinas/uso terapéutico , Daño por Reperfusión Miocárdica/patología , Miocardio/enzimología , Miocardio/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Proteínas Recombinantes/uso terapéutico
12.
PLoS One ; 10(6): e0129288, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26068620

RESUMEN

The cell surface proteoglycan syndecan-4 has been reported to be crucial for muscle differentiation, but the molecular mechanisms still remain to be fully understood. During in vitro differentiation of bovine muscle cells immunocytochemical analyses showed strong labelling of syndecan-4 intracellularly, in close proximity with Golgi structures, in membranes of intracellular vesicles and finally, in the nuclear area including the nuclear envelope. Chase experiments showed that syndecan-4 was internalized from the plasma membrane during this process. Furthermore, when syndecan-4 was knocked down by siRNA more myotubes were formed, and the expression of myogenic transcription factors, ß1-integrin and actin was influenced. However, when bovine muscle cells were treated with a cell-penetrating peptide containing the cytoplasmic region of syndecan-4, myoblast fusion and thus myotube formation was blocked, both in normal cells and in syndecan-4 knock down cells. Altogether this suggests that the cytoplasmic domain of syndecan-4 is important in regulation of myogenesis. The internalization of syndecan-4 from the plasma membrane during muscle differentiation and the nuclear localization of syndecan-4 in differentiated muscle cells may be part of this regulation, and is a novel aspect of syndecan biology which merits further studies.


Asunto(s)
Diferenciación Celular , Membrana Celular/metabolismo , Desarrollo de Músculos , Fibras Musculares Esqueléticas/metabolismo , Sindecano-4/metabolismo , Secuencia de Aminoácidos , Animales , Bovinos , Células Cultivadas , Datos de Secuencia Molecular , Fibras Musculares Esqueléticas/citología , Estructura Terciaria de Proteína , Transporte de Proteínas , Sindecano-4/química , Sindecano-4/genética
13.
Membranes (Basel) ; 4(3): 424-46, 2014 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-25102001

RESUMEN

The EGFR- or ErbB-family of receptor tyrosine kinases consists of EGFR/ErbB1, ErbB2/HER2, ErbB3/HER3 and ErbB4/HER4. Receptor activation and downstream signaling are generally initiated upon ligand-induced receptor homo- or heterodimerization at the plasma membrane, and endocytosis and intracellular membrane transport are crucial for regulation of the signaling outcome. Among the receptors, ErbB2 is special in several ways. Unlike the others, ErbB2 has no known ligand, but is still the favored dimerization partner. Furthermore, while the other receptors are down-regulated either constitutively or upon ligand-binding, ErbB2 is resistant to down-regulation, and also inhibits down-regulation of its partner upon heterodimerization. The reason(s) why ErbB2 is resistant to down-regulation are the subject of debate. Contrary to other ErbB-proteins, mature ErbB2 needs Hsp90 as chaperone. Several data suggest that Hsp90 is an important regulator of factors like ErbB2 stability, dimerization and/or signaling. Hsp90 inhibitors induce degradation of ErbB2, but whether Hsp90 directly makes ErbB2 endocytosis resistant is unclear. Exposure to anti-ErbB2 antibodies can also induce down-regulation of ErbB2. Down-regulation induced by Hsp90 inhibitors or antibodies does at least partly involve internalization and endosomal sorting to lysosomes for degradation, but also retrograde trafficking to the nucleus has been reported. In this review, we will discuss different molecular mechanisms suggested to be important for making ErbB2 resistant to down-regulation, and review how membrane trafficking is involved when down-regulation and/or relocalization of ErbB2 is induced.

14.
Carcinogenesis ; 34(9): 2031-8, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23698633

RESUMEN

Overexpression of ErbB2 and ErbB3 is found in several human cancers, and ErbB2-ErbB3 heterodimers are known as the most potent signaling units among ErbB dimers. While ErbB2 probably undergoes weak endocytosis, ErbB3 is readily internalized even in the absence of added ligand and without requirement for kinase activity. Overexpression of ErbB2 has been demonstrated to inhibit epidermal growth factor-induced internalization and degradation of epidermal growth factor receptor. This happens due to epidermal growth factor receptor-ErbB2 dimerization and can be counteracted by the anti-ErbB2 antibody pertuzumab, which binds the dimerization arm of ErbB2. Pertuzumab does also inhibit ErbB2-ErbB3 dimerization, but to what extent this has effect on constitutive and/or ligand-induced downregulation of ErbB3 is not known. In this study, we demonstrate that expression of ErbB2 as such did not block constitutive internalization of ErbB3, but that heregulin-induced degradation of ErbB3 was significantly slowed in cells expressing high levels of ErbB2. Incubation with pertuzumab did, however, counteract this effect. This indicates that the formation of ErbB2-ErbB3 heterodimers inhibits downregulation of ErbB3 and supports the notion that pertuzumab inhibits ErbB2 dimerization. The inhibitory effect of pertuzumab on ligand-induced ErbB2-ErbB3 heterodimerization was confirmed by the observation that pertuzumab inhibited heregulin-induced phosphorylation of ErbB3 in cells expressing ErbB2 and efficiently reduced heregulin-induced downstream signaling in cells expressing low levels of ErbB2. Altogether the results indicate that pertuzumab can be a valuable therapeutic agent not only in cancers overexpressing ErbB2 but also in cancers co-expressing ErbB2 and ErbB3.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Neoplasias de la Mama/genética , Receptor ErbB-2/metabolismo , Receptor ErbB-3/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Dimerización , Regulación hacia Abajo , Endocitosis/efectos de los fármacos , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ligandos , Células MCF-7 , Fosforilación , Unión Proteica , Proteolisis/efectos de los fármacos , Receptor ErbB-2/química , Receptor ErbB-3/química , Transducción de Señal/efectos de los fármacos
15.
Exp Cell Res ; 319(3): 32-45, 2013 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-23127513

RESUMEN

The oncoprotein ErbB2 is endocytosis-deficient, probably due to its interaction with Heat shock protein 90. We previously demonstrated that clathrin-dependent endocytosis of ErbB2 is induced upon incubation of cells with Ansamycin derivatives, such as geldanamycin and its derivative 17-AAG. Furthermore, we have previously demonstrated that a preubiquitinated chimeric EGFR (EGFR-Ub(4)) is constitutively endocytosed in a clathrin-dependent manner. We now demonstrate that also an ErbB2-Ub(4) chimera is endocytosed constitutively and clathrin-dependently. Upon expression, the ErbB2-Ub(4) was further ubiquitinated, and by Western blotting, we demonstrated the formation of both Lys48-linked and Lys63-linked polyubiquitin chains. ErbB2-Ub(4) was constitutively internalized and eventually sorted to late endosomes and lysosomes where the fusion protein was degraded. ErbB2-Ub(4) was not cleaved prior to internalization. Interestingly, over-expression of Ubiquitin Interaction Motif-containing dominant negative fragments of the clathrin adaptor proteins epsin1 and Eps15 negatively affected endocytosis of ErbB2. Altogether, this argues that ubiquitination is sufficient to induce clathrin-mediated endocytosis and lysosomal degradation of the otherwise plasma membrane localized ErbB2. Also, it appears that C-terminal cleavage is not required for endocytosis.


Asunto(s)
Endocitosis/fisiología , Lisosomas/metabolismo , Proteolisis , Receptor ErbB-2/metabolismo , Ubiquitinación/fisiología , Animales , Benzoquinonas/farmacología , Membrana Celular/metabolismo , Células Cultivadas , Endocitosis/efectos de los fármacos , Lactamas Macrocíclicas/farmacología , Lisosomas/efectos de los fármacos , Modelos Biológicos , Inhibidores de Proteínas Quinasas/farmacología , Multimerización de Proteína , Proteolisis/efectos de los fármacos , Receptor ErbB-2/química , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Porcinos , Ubiquitina/metabolismo
16.
Exp Cell Res ; 318(20): 2578-91, 2012 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-22975728

RESUMEN

The monoclonal antibody C225 (Cetuximab) blocks binding of ligand to the epidermal growth factor receptor (EGFR). In addition, it is known that incubation with C225 induces endocytosis of the EGFR. This endocytosis has previously been shown to be increased when C225 is combined with an additional monoclonal anti-EGFR antibody. However, the effects of antibody combinations on EGFR activation, endocytosis, trafficking and degradation have been unclear. By binding a secondary antibody to the C225-EGFR complex, we here demonstrate that a combination of antibodies can efficiently internalize and degrade the EGFR. Although the combination of antibodies activated the EGFR kinase and induced ubiquitination of the EGFR, the kinase activity was not required for internalization of the EGFR. In contrast to EGF-induced EGFR down-regulation, the antibody combination efficiently degraded the EGFR without initiating downstream proliferative signaling. The antibody-induced internalization of EGFR was found not to depend on clathrin and/or dynamin, but depended on actin polymerization, suggesting induction of macropinocytosis. Macropinocytosis may cause internalization of large membrane areas, and this could explain the highly efficient internalization of the EGFR induced by combination of antibodies.


Asunto(s)
Anticuerpos Antiidiotipos/farmacología , Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Regulación hacia Abajo/inmunología , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/biosíntesis , Inmunoglobulina G/inmunología , Pinocitosis/inmunología , Animales , Anticuerpos Monoclonales Humanizados , Cetuximab , Quimioterapia Combinada , Células Endoteliales/citología , Células Endoteliales/metabolismo , Receptores ErbB/genética , Humanos , Ratones , Porcinos
17.
PLoS One ; 7(7): e40673, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22815786

RESUMEN

BACKGROUND: In addition to lowering cholesterol, statins are thought to beneficially modulate inflammation. Several chemokines including CXCL1/growth-related oncogene (GRO)-α, CXCL8/interleukin (IL)-8 and CCL2/monocyte chemoattractant protein (MCP)-1 are important in the pathogenesis of atherosclerosis and can be influenced by statin-treatment. Recently, we observed that atorvastatin-treatment alters the intracellular content and subcellular distribution of GRO-α in cultured human umbilical vein endothelial cells (HUVECs). The objective of this study was to investigate the mechanisms involved in this phenomenon. METHODOLOGY/ PRINCIPAL FINDINGS: The effect of atorvastatin on secretion levels and subcellular distribution of GRO-α, IL-8 and MCP-1 in HUVECs activated by interleukin (IL)-1ß were evaluated by ELISA, confocal microscopy and immunoelectron microscopy. Atorvastatin increased the intracellular contents of GRO-α, IL-8, and MCP-1 and induced colocalization with E-selectin in multivesicular bodies. This effect was prevented by adding the isoprenylation substrate GGPP, but not the cholesterol precursor squalene, indicating that atorvastatin exerts these effects by inhibiting isoprenylation rather than depleting the cells of cholesterol. CONCLUSIONS/ SIGNIFICANCE: Atorvastatin targets inflammatory chemokines to the endocytic pathway and multivesicular bodies and may contribute to explain the anti-inflammatory effect of statins at the level of endothelial cell function.


Asunto(s)
Quimiocinas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/citología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Cuerpos Multivesiculares/metabolismo , Atorvastatina , Compartimento Celular/efectos de los fármacos , Quimiocina CXCL1/metabolismo , Selectina E/metabolismo , Endocitosis/efectos de los fármacos , Ácidos Grasos Monoinsaturados/farmacología , Fluvastatina , Ácidos Heptanoicos/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/ultraestructura , Humanos , Indoles/farmacología , Interleucina-1beta/farmacología , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Cuerpos Multivesiculares/efectos de los fármacos , Pravastatina/farmacología , Prenilación/efectos de los fármacos , Pirroles/farmacología , Simvastatina/farmacología , Solubilidad , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Tetraspanina 30/metabolismo
18.
Carcinogenesis ; 33(5): 1031-9, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22436610

RESUMEN

The oncoprotein ErbB3 is overexpressed in several human cancers, for example in pancreatic adenocarcinoma and in ovarian cancers, and ErbB3-containing heterodimers have been demonstrated to be potent signaling units in carcinogenesis. This especially applies to ErbB2-ErbB3 and epidermal growth factor receptor (EGFR)-ErbB3 heterodimers providing anti-apoptotic signaling. Relatively little is understood about the signaling of EGFR-ErbB3 heterodimers and especially about mechanisms involved in downregulation of ErbB3 from the plasma membrane. This is in contrast to EGFR homodimers, for which trafficking has been extensively characterized. In the present study, we have investigated mechanisms involved in endocytosis of ErbB3 in porcine aortic endothelial cells stably expressing either ErbB3 only or stably expressing ErbB3 and EGFR. Our data show that ErbB3 is endocytosed in the absence of added ligand, independently of its tyrosine phosphorylation state and in a clathrin-dependent manner. Functional EGFR-ErbB3 heterodimers were observed to be formed, and dimerization with ErbB3 was observed to negatively affect endocytosis of the EGFR.


Asunto(s)
Clatrina/metabolismo , Proteínas Oncogénicas/metabolismo , Receptor ErbB-3/metabolismo , Animales , Apoptosis/fisiología , Membrana Celular/metabolismo , Dimerización , Endocitosis , Células Endoteliales/metabolismo , Receptores ErbB/metabolismo , Células HeLa , Hemoproteínas/metabolismo , Humanos , Ligandos , Fosforilación , Receptor ErbB-2/metabolismo , Transducción de Señal/fisiología , Porcinos , Células Tumorales Cultivadas
19.
Pharmaceuticals (Basel) ; 5(7): 674-89, 2012 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-24281706

RESUMEN

ErbB2 is an important oncogenic protein involved in carcinogenesis of, among others, breast, gastric, and ovarian carcinoma. Over-expression of ErbB2 is found in almost 20% of breast cancers, and this results in proliferative and anti-apoptotic signalling. ErbB2 is therefore an important treatment target. Antibodies recognizing full-length ErbB2 are clinically established, and drugs targeting the ErbB2 stabilizing heat shock protein 90 (Hsp90) are under clinical evaluation. We have investigated effects of the ErbB2-binding antibodies trastuzumab and pertuzumab alone and in combination, as well as the effect of the antibodies in combination with the Hsp90 inhibitor 17-AAG. Our results confirm the notion that combination of different ErbB2-binding antibodies more efficiently down-regulates ErbB2 than does one antibody in isolation. Additionally, our data demonstrate that ErbB2 is most efficiently down-regulated upon incubation with anti-ErbB2 antibodies in combination with Hsp90 inhibitors. The combination of anti-ErbB2 antibodies, and especially the combination of antibodies with 17-AAG, did also increase the inhibition of Akt activation of either agent, which could suggest an anti-proliferative effect. In such case, combining these agents could be beneficial in treatment of tumors not responding to trastuzumab only.

20.
J Immunother ; 34(7): 550-5, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21760527

RESUMEN

The epidermal growth factor receptor (EGFR) is overexpressed in several epithelial tumors such as breast, ovarian, and colon cancers. Nimotuzumab and Cetuximab are antibodies that inhibit ligand binding upon interaction with the EGFR, thereby indirectly inactivating the EGFR kinase. The ability of an antibody to counteract growth depends on its mechanism of action as well as on its binding affinity. Nimotuzumab has lower binding affinity for the EGFR than does Cetuximab. In addition, a mechanistic difference has recently been suggested to explain the different clinical effects of Nimotuzumab and Cetuximab, arguing that Nimotuzumab partly permits kinase activity and downstream signaling under conditions where binding of EGF is inhibited. We have in the current study compared the effects of Nimotuzumab and Cetuximab on binding of EGF as well as on inhibition of constitutive EGFR-ErbB2 dimerization and downstream activation of Erk. Our results demonstrate that (at least in EGFR-overexpressing cells), in contrast to the recently published mechanistic model, Nimotuzumab not only inhibits EGF-stimulated, but also ligand-independent (basal) signaling although at higher concentrations than required with Cetuximab.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales/farmacología , Receptores ErbB/metabolismo , Transducción de Señal/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cetuximab , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/inmunología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Glandulares y Epiteliales/inmunología , Multimerización de Proteína/efectos de los fármacos , Receptor ErbB-2/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...