Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Sci ; 135(5)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34981808

RESUMEN

High-density lipoproteins (HDLs) prevent cell death induced by a variety of cytotoxic drugs. The underlying mechanisms are however still poorly understood. Here, we present evidence that HDLs efficiently protect cells against thapsigargin (TG), a sarco/endoplasmic reticulum (ER) Ca2+-ATPase (SERCA) inhibitor, by extracting the drug from cells. Drug efflux could also be triggered to some extent by low-density lipoproteins and serum. HDLs did not reverse the non-lethal mild ER stress response induced by low TG concentrations or by SERCA knockdown, but HDLs inhibited the toxic SERCA-independent effects mediated by high TG concentrations. HDLs could extract other lipophilic compounds, but not hydrophilic substances. This work shows that HDLs utilize their capacity of loading themselves with lipophilic compounds, akin to their ability to extract cellular cholesterol, to reduce the cell content of hydrophobic drugs. This can be beneficial if lipophilic xenobiotics are toxic but may be detrimental to the therapeutic benefit of lipophilic drugs such as glibenclamide.


Asunto(s)
Lipoproteínas HDL , Preparaciones Farmacéuticas , Calcio/metabolismo , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Tapsigargina/farmacología
2.
Proc Natl Acad Sci U S A ; 117(50): 31871-31881, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33257567

RESUMEN

TAT-RasGAP317-326 is a cell-penetrating peptide-based construct with anticancer and antimicrobial activities. This peptide kills a subset of cancer cells in a manner that does not involve known programmed cell death pathways. Here we have elucidated the mode of action allowing TAT-RasGAP317-326 to kill cells. This peptide binds and disrupts artificial membranes containing lipids typically enriched in the inner leaflet of the plasma membrane, such as phosphatidylinositol-bisphosphate (PIP2) and phosphatidylserine (PS). Decreasing the amounts of PIP2 in cells renders them more resistant to TAT-RasGAP317-326, while reducing the ability of cells to repair their plasma membrane makes them more sensitive to the peptide. The W317A TAT-RasGAP317-326 point mutant, known to have impaired killing activities, has reduced abilities to bind and permeabilize PIP2- and PS-containing membranes and to translocate through biomembranes, presumably because of a higher propensity to adopt an α-helical state. This work shows that TAT-RasGAP317-326 kills cells via a form of necrosis that relies on the physical disruption of the plasma membrane once the peptide targets specific phospholipids found on the cytosolic side of the plasma membrane.


Asunto(s)
Muerte Celular/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Proteínas Activadoras de GTPasa/farmacología , Fragmentos de Péptidos/farmacología , Fosfatidilinositol 4,5-Difosfato/metabolismo , Fosfatidilserinas/metabolismo , Animales , Células CHO , Membrana Celular/metabolismo , Membrana Celular/ultraestructura , Cricetulus , Proteínas Activadoras de GTPasa/uso terapéutico , Células HeLa , Humanos , Liposomas/metabolismo , Liposomas/ultraestructura , Microscopía Electrónica , Simulación de Dinámica Molecular , Neoplasias/tratamiento farmacológico , Resonancia Magnética Nuclear Biomolecular , Fragmentos de Péptidos/uso terapéutico
3.
J Virol ; 93(6)2019 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-30626681

RESUMEN

Arenaviruses are a large family of emerging enveloped negative-strand RNA viruses that include several causative agents of viral hemorrhagic fevers. For cell entry, human-pathogenic arenaviruses use different cellular receptors and endocytic pathways that converge at the level of acidified late endosomes, where the viral envelope glycoprotein mediates membrane fusion. Inhibitors of arenavirus entry hold promise for therapeutic antiviral intervention and the identification of "druggable" targets is of high priority. Using a recombinant vesicular stomatitis virus pseudotype platform, we identified the clotrimazole-derivative TRAM-34, a highly selective antagonist of the calcium-activated potassium channel KCa3.1, as a specific entry inhibitor for arenaviruses. TRAM-34 specifically blocked entry of most arenaviruses, including hemorrhagic fever viruses, but not Lassa virus and other enveloped viruses. Anti-arenaviral activity was likewise observed with the parental compound clotrimazole and the derivative senicapoc, whereas structurally unrelated KCa3.1 inhibitors showed no antiviral effect. Deletion of KCa3.1 by CRISPR/Cas9 technology did not affect the antiarenaviral effect of TRAM-34, indicating that the observed antiviral effect of clotrimazoles was independent of the known pharmacological target. The drug affected neither virus-cell attachment, nor endocytosis, suggesting an effect on later entry steps. Employing a quantitative cell-cell fusion assay that bypasses endocytosis, we demonstrate that TRAM-34 specifically inhibits arenavirus-mediated membrane fusion. In sum, we uncover a novel antiarenaviral action of clotrimazoles that currently undergo in vivo evaluation in the context of other human diseases. Their favorable in vivo toxicity profiles and stability opens the possibility to repurpose clotrimazole derivatives for therapeutic intervention against human-pathogenic arenaviruses.IMPORTANCE Emerging human-pathogenic arenaviruses are causative agents of severe hemorrhagic fevers with high mortality and represent serious public health problems. The current lack of a licensed vaccine and the limited treatment options makes the development of novel antiarenaviral therapeutics an urgent need. Using a recombinant pseudotype platform, we uncovered that clotrimazole drugs, in particular TRAM-34, specifically inhibit cell entry of a range of arenaviruses, including important emerging human pathogens, with the exception of Lassa virus. The antiviral effect was independent of the known pharmacological drug target and involved inhibition of the unusual membrane fusion mechanism of arenaviruses. TRAM-34 and its derivatives currently undergo evaluation against a number of human diseases and show favorable toxicity profiles and high stability in vivo Our study provides the basis for further evaluation of clotrimazole derivatives as antiviral drug candidates. Their advanced stage of drug development will facilitate repurposing for therapeutic intervention against human-pathogenic arenaviruses.


Asunto(s)
Antivirales/farmacología , Arenavirus/efectos de los fármacos , Clotrimazol/farmacología , Fusión de Membrana/efectos de los fármacos , Células A549 , Animales , Infecciones por Arenaviridae/tratamiento farmacológico , Línea Celular , Línea Celular Tumoral , Chlorocebus aethiops , Endocitosis/efectos de los fármacos , Células HEK293 , Células HeLa , Fiebres Hemorrágicas Virales/tratamiento farmacológico , Fiebres Hemorrágicas Virales/virología , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Virus Lassa/efectos de los fármacos , Células Vero , Proteínas del Envoltorio Viral/metabolismo , Acoplamiento Viral/efectos de los fármacos , Internalización del Virus/efectos de los fármacos
4.
J Cell Sci ; 128(18): 3502-13, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26224876

RESUMEN

The nuclear factor κB (NF-κB) transcription factor is a master regulator of inflammation. Short-term NF-κB activation is generally beneficial. However, sustained NF-κB might be detrimental, directly causing apoptosis of cells or leading to a persistent damaging inflammatory response. NF-κB activity in stressed cells needs therefore to be controlled for homeostasis maintenance. In mildly stressed cells, caspase-3 cleaves p120 RasGAP, also known as RASA1, into an N-terminal fragment, which we call fragment N. We show here that this fragment is a potent NF-κB inhibitor. Fragment N decreases the transcriptional activity of NF-κB by promoting its export from the nucleus. Cells unable to generate fragment N displayed increased NF-κB activation upon stress. Knock-in mice expressing an uncleavable p120 RasGAP mutant showed exaggerated NF-κB activation when their epidermis was treated with anthralin, a drug used for the treatment of psoriasis. Our study provides biochemical and genetic evidence of the importance of the caspase-3-p120-RasGAP stress-sensing module in the control of stress-induced NF-κB activation.


Asunto(s)
Caspasa 3/metabolismo , FN-kappa B/metabolismo , Fragmentos de Péptidos , Proteína Activadora de GTPasa p120/metabolismo , Animales , Células HEK293 , Humanos , Ratones , Ratones Noqueados , FN-kappa B/química , Ratas , Estrés Fisiológico/fisiología , Proteína Activadora de GTPasa p120/química
5.
Mol Carcinog ; 56(6): 1680-1684, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28150874

RESUMEN

Activation of oncogenes is the initial step in cellular transformation. Oncogenes favor aberrant proliferation, which, at least initially, induces cellular stress. This oncogenic stress can act as a safeguard mechanism against further transformation by inducing senescence or apoptosis. Yet, the few premalignant cells that tolerate and escape these senescent or apoptotic responses are those that will ultimately generate tumors. The caspase-3/p120 RasGAP module is a stress-sensing device that promotes survival under mild stress conditions. A point mutation in RasGAP that prevents its cleavage by caspase-3 inactivates the pro-survival capacity of the device. When the mice homozygous for this mutation (D455A knock-in mice) are patho-physiologically challenged, they experience much stronger cellular damage than their wild-type counterparts and the affected organs rapidly lose their functionality. We reasoned that the caspase-3/p120 RasGAP module could help premalignant cells to cope with oncogenic stress and hence favor the development of tumors. Using gamma-irradiation and N-ethyl-N-nitrosourea (ENU) as tumor initiators, we assessed the survival advantage that the caspase-3/p120 RasGAP module could provide to premalignant cells. No difference in overall mortality between wild-type and D455A knock-in mice were observed. However, the number of ENU-induced liver tumors in the knock-in mice was higher than in control mice. These results indicate that the caspase-3/p120 RasGAP stress-sensing module impacts on carcinogen-induced liver cancer incidence but not sufficiently so as to affect overall survival. Hence, gamma irradiation and ENU-induced tumorigenesis processes do not critically rely on a survival mechanism that contributes to the maintenance of organ homeostasis in stressed healthy tissues.


Asunto(s)
Caspasa 3/metabolismo , Neoplasias Hepáticas/metabolismo , Hígado/patología , Neoplasias Inducidas por Radiación/metabolismo , Proteína Activadora de GTPasa p120/metabolismo , Animales , Línea Celular , Etilnitrosourea , Rayos gamma , Incidencia , Hígado/metabolismo , Hígado/efectos de la radiación , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos C57BL , Neoplasias Inducidas por Radiación/patología
6.
J Biol Chem ; 290(32): 19653-65, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26109071

RESUMEN

Fibroblast growth factor receptors (FGFRs) are involved in proliferative and differentiation physiological responses. Deregulation of FGFR-mediated signaling involving the Ras/PI3K/Akt and the Ras/Raf/ERK MAPK pathways is causally involved in the development of several cancers. The caspase-3/p120 RasGAP module is a stress sensor switch. Under mild stress conditions, RasGAP is cleaved by caspase-3 at position 455. The resulting N-terminal fragment, called fragment N, stimulates anti-death signaling. When caspase-3 activity further increases, fragment N is cleaved at position 157. This generates a fragment, called N2, that no longer protects cells. Here, we investigated in Xenopus oocytes the impact of RasGAP and its fragments on FGF1-mediated signaling during G2/M cell cycle transition. RasGAP used its N-terminal Src homology 2 domain to bind FGFR once stimulated by FGF1, and this was necessary for the recruitment of Akt to the FGFR complex. Fragment N, which did not associate with the FGFR complex, favored FGF1-induced ERK stimulation, leading to accelerated G2/M transition. In contrast, fragment N2 bound the FGFR, and this inhibited mTORC2-dependent Akt Ser-473 phosphorylation and ERK2 phosphorylation but not phosphorylation of Akt on Thr-308. This also blocked cell cycle progression. Inhibition of Akt Ser-473 phosphorylation and entry into G2/M was relieved by PHLPP phosphatase inhibition. Hence, full-length RasGAP favors Akt activity by shielding it from deactivating phosphatases. This shielding was abrogated by fragment N2. These results highlight the role played by RasGAP in FGFR signaling and how graded stress intensities, by generating different RasGAP fragments, can positively or negatively impact this signaling.


Asunto(s)
Caspasa 3/metabolismo , Factor 1 de Crecimiento de Fibroblastos/metabolismo , Oocitos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteína Activadora de GTPasa p120/metabolismo , Animales , Caspasa 3/genética , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Femenino , Factor 1 de Crecimiento de Fibroblastos/genética , Factor 1 de Crecimiento de Fibroblastos/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Regulación de la Expresión Génica , Humanos , Diana Mecanicista del Complejo 2 de la Rapamicina , Microinyecciones , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oocitos/citología , Oocitos/efectos de los fármacos , Ovario/citología , Ovario/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , Cultivo Primario de Células , Estructura Terciaria de Proteína , Proteolisis , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Xenopus laevis , Proteína Activadora de GTPasa p120/genética
7.
BMC Cancer ; 16(1): 781, 2016 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-27724856

RESUMEN

BACKGROUND: The successful targeting of neuroblastoma (NB) by associating tumor-initiating cells (TICs) is a major challenge in the development of new therapeutic strategies. The subfamily of aldehyde dehydrogenases 1 (ALDH1) isoenzymes, which comprises ALDH1A1, ALDH1A2, and ALDH1A3, is involved in the synthesis of retinoic acid, and has been identified as functional stem cell markers in diverse cancers. By combining serial neurosphere passages with gene expression profiling, we have previously identified ALDH1A2 and ALDH1A3 as potential NB TICs markers in patient-derived xenograft tumors. In this study, we explored the involvement of ALDH1 isoenzymes and the related ALDH activity in NB aggressive properties. METHODS: ALDH activity and ALDH1A1/A2/A3 expression levels were measured using the ALDEFLUOR™ kit, and by real-time PCR, respectively. ALDH activity was inhibited using the specific ALDH inhibitor diethylaminobenzaldehyde (DEAB), and ALDH1A3 gene knock-out was generated through the CRISPR/Cas9 technology. RESULTS: We first confirmed the enrichment of ALDH1A2 and ALDH1A3 mRNA expression in NB cell lines and patient-derived xenograft tumors during neurosphere passages. We found that high ALDH1A1 expression was associated with less aggressive NB tumors and cell lines, and correlated with favorable prognostic factors. In contrast, we observed that ALDH1A3 was more widely expressed in NB cell lines and was associated with poor survival and high-risk prognostic factors. We also identified an important ALDH activity in various NB cell lines and patient-derived xenograft tumors. Specific inhibition of ALDH activity with diethylaminobenzaldehyde (DEAB) resulted in a strong reduction of NB cell clonogenicity, and TIC self-renewal potential, and partially enhanced NB cells sensitivity to 4-hydroxycyclophosphamide. Finally, the specific knock-out of ALDH1A3 via CRISPR/Cas9 gene editing reduced NB cell clonogenicity, and mediated a cell type-dependent inhibition of TIC self-renewal properties. CONCLUSIONS: Together our data uncover the participation of ALDH enzymatic activity in the aggressive properties and 4-hydroxycyclophosphamide resistance of NB, and show that the specific ALDH1A3 isoenzyme increases the aggressive capacities of a subset of NB cells.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Neuroblastoma/diagnóstico , Neuroblastoma/enzimología , Fenotipo , Aldehído Deshidrogenasa/genética , Familia de Aldehído Deshidrogenasa 1 , Aldehído Oxidorreductasas/genética , Aldehído Oxidorreductasas/metabolismo , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Activación Enzimática , Expresión Génica , Técnicas de Inactivación de Genes , Xenoinjertos , Humanos , Isoenzimas , Ratones , Neuroblastoma/genética , Pronóstico , Retinal-Deshidrogenasa/genética , Retinal-Deshidrogenasa/metabolismo , Transcriptoma
9.
J Biol Chem ; 289(34): 23701-11, 2014 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-25008324

RESUMEN

TAT-RasGAP317-326, a cell-permeable 10-amino acid-long peptide derived from the N2 fragment of p120 Ras GTPase-activating protein (RasGAP), sensitizes tumor cells to apoptosis induced by various anticancer therapies. This RasGAP-derived peptide, by targeting the deleted in liver cancer-1 (DLC1) tumor suppressor, also hampers cell migration and invasion by promoting cell adherence and by inhibiting cell movement. Here, we systematically investigated the role of each amino acid within the RasGAP317-326 sequence for the anticancer activities of TAT-RasGAP317-326. We report here that the first three amino acids of this sequence, tryptophan, methionine, and tryptophan (WMW), are necessary and sufficient to sensitize cancer cells to cisplatin-induced apoptosis and to reduce cell migration. The WMW motif was found to be critical for the binding of fragment N2 to DLC1. These results define the interaction mode between the active anticancer sequence of RasGAP and DLC1. This knowledge will facilitate the design of small molecules bearing the tumor-sensitizing and antimetastatic activities of TAT-RasGAP317-326.


Asunto(s)
Secuencias de Aminoácidos , Antineoplásicos/farmacología , Proteínas Activadoras de GTPasa/farmacología , Fragmentos de Péptidos/farmacología , Secuencia de Aminoácidos , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Secuencia de Bases , Calorimetría , Línea Celular Tumoral , Cartilla de ADN , Proteínas Activadoras de GTPasa/química , Células HEK293 , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Reacción en Cadena de la Polimerasa , Relación Estructura-Actividad
10.
Cancer Metastasis Rev ; 33(1): 87-100, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24338004

RESUMEN

Metastases are responsible for most cancer-related deaths. One of the hallmarks of metastatic cells is increased motility and migration through extracellular matrixes. These processes rely on specific small GTPases, in particular those of the Rho family. Deleted in liver cancer-1 (DLC1) is a tumor suppressor that bears a RhoGAP activity. This protein is lost in most cancers, allowing malignant cells to proliferate and disseminate in a Rho-dependent manner. However, DLC1 is also a scaffold protein involved in alternative pathways leading to tumor and metastasis suppressor activities. Recently, substantial information has been gathered on these mechanisms and this review is aiming at describing the potential and known alternative GAP-independent mechanisms allowing DLC1 to impair migration, invasion, and metastasis formation.


Asunto(s)
Proteínas Activadoras de GTPasa/genética , Neoplasias/genética , Proteínas Supresoras de Tumor/genética , Movimiento Celular/genética , Proteínas Activadoras de GTPasa/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Modelos Genéticos , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Supresoras de Tumor/metabolismo
11.
Handb Exp Pharmacol ; 224: 405-21, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25522996

RESUMEN

The prevalence of type 2 diabetes mellitus and of the metabolic syndrome is rising worldwide and reaching epidemic proportions. These pathologies are associated with significant morbidity and mortality, in particular with an excess of cardiovascular deaths. Type 2 diabetes mellitus and the cluster of pathologies including insulin resistance, central obesity, high blood pressure, and hypertriglyceridemia that constitute the metabolic syndrome are associated with low levels of HDL cholesterol and the presence of dysfunctional HDLs. We here review the epidemiological evidence and the potential underlying mechanisms of this association. We first discuss the well-established association of type 2 diabetes mellitus and insulin resistance with alterations of lipid metabolism and how these alterations may lead to low levels of HDL cholesterol and the occurrence of dysfunctional HDLs. We then present and discuss the evidence showing that HDL modulates insulin sensitivity, insulin-independent glucose uptake, insulin secretion, and beta cell survival. A dysfunction in these actions could play a direct role in the pathogenesis of type 2 diabetes mellitus.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Lipoproteínas HDL/metabolismo , Síndrome Metabólico/metabolismo , Animales , Biomarcadores/sangre , HDL-Colesterol/sangre , Diabetes Mellitus Tipo 2/sangre , Humanos , Células Secretoras de Insulina/metabolismo , Lipoproteínas HDL/sangre , Síndrome Metabólico/sangre , Factores Protectores , Medición de Riesgo , Factores de Riesgo
13.
Int J Cancer ; 135(1): 242-7, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24347041

RESUMEN

The p120 RasGAP protein negatively regulates Ras via its GAP domain. RasGAP carries several other domains that modulate several signaling molecules such as Rho. RasGAP is also a caspase-3 substrate. One of the caspase-3-generated RasGAP fragments, corresponding to amino acids 158-455 and called fragment N2, was previously reported to specifically sensitize cancer cells to death induced by various anticancer agents. Here, we show that fragment N2 inhibits migration in vitro and that it impairs metastatic progression of breast cancer to the lung. Hence, stress-activated caspase-3 might contribute to the suppression of metastasis through the generation of fragment N2. These results indicate that the activity borne by fragment N2 has a potential therapeutic relevance to counteract the metastatic process.


Asunto(s)
Caspasa 3/genética , Movimiento Celular/genética , Fragmentos de Péptidos/genética , Proteínas Activadoras de ras GTPasa/genética , Apoptosis/genética , Neoplasias de la Mama , Caspasa 3/química , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis de la Neoplasia/genética , Fragmentos de Péptidos/química , Transfección
14.
Apoptosis ; 19(4): 719-33, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24362790

RESUMEN

The increase of cancer specificity and efficacy of anti-tumoral agents are prime strategies to overcome the deleterious side effects associated with anti-cancer treatments. We described earlier a cell-permeable protease-resistant peptide derived from the p120 RasGAP protein, called TAT-RasGAP317-326, as being an efficient tumor-specific sensitizer to apoptosis induced by genotoxins in vitro and in vivo. Bcl-2 family members regulate the intrinsic apoptotic response and as such could be targeted by TAT-RasGAP317-326. Our results indicate that the RasGAP-derived peptide increases cisplatin-induced Bax activation. We found no evidence, using in particular knock-out cells, of an involvement of other Bcl-2 family proteins in the tumor-specific sensitization activity of TAT-RasGAP317-326. The absence of Bax and Bak in mouse embryonic fibroblasts rendered them resistant to cisplatin-induced apoptosis and consequently to the sensitizing action of the RasGAP-derived peptide. Surprisingly, in the HCT116 colon carcinoma cell line, the absence of Bax and Bak did not prevent cisplatin-induced apoptosis and the ability of TAT-RasGAP317-326 to augment this response. Our study also revealed that p53, while required for an efficient genotoxin-induced apoptotic response, is dispensable for the ability of the RasGAP-derived peptide to improve the capacity of genotoxins to decrease long-term survival of cancer cells. Hence, even though genotoxin-induced Bax activity can be increased by TAT-RasGAP317-326, the sensitizing activity of the RasGAP-derived peptide can operate in the absence of a functional mitochondrial intrinsic death pathway.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteínas Activadoras de GTPasa/farmacología , Fragmentos de Péptidos/farmacología , Proteína Destructora del Antagonista Homólogo bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Animales , Línea Celular Tumoral/efectos de los fármacos , Permeabilidad de la Membrana Celular , Cisplatino/farmacología , Humanos , Ratones Noqueados , Proteína p53 Supresora de Tumor/metabolismo
17.
Eur J Pharm Biopharm ; 184: 116-124, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36709921

RESUMEN

Cell-penetrating peptides (CPPs) are short (<30 amino acids), generally cationic, peptides that deliver diverse cargos into cells. CPPs access the cytosol either by direct translocation through the plasma membrane or via endocytosis followed by endosomal escape. Both direct translocation and endosomal escape can occur simultaneously, making it non-trivial to specifically study endosomal escape alone. Here we depolarize the plasma membrane and showed that it inhibits the direct translocation of several CPPs but does not affect their uptake into endosomes. Despite good endocytic uptake many CPPs previously considered to access the cytosol via endosomal escape, failed to access the cytosol once direct translocation was abrogated. Even CPPs designed for enhanced endosomal escape actually showed negligible endosomal escape into the cytosol. Our data reveal that cytosolic localization of CPPs occurs mainly by direct translocation across the plasma membrane. Cell depolarization represents a simple manipulation to stringently test the endosomal escape capacity of CPPs.


Asunto(s)
Péptidos de Penetración Celular , Péptidos de Penetración Celular/química , Endosomas/metabolismo , Endocitosis , Transporte Biológico , Membrana Celular/metabolismo
18.
J Mol Biol ; 435(4): 167933, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36581244

RESUMEN

Native molecular weight (MW) is one of the defining features of proteins. Denaturing gel electrophoresis (SDS-PAGE) is a very popular technique for separating proteins and determining their MW. Coupled with antibody-based detection, SDS-PAGE is widely applied for protein identification and quantitation. Yet, electrophoresis is poorly reproducible and the MWs obtained are often inaccurate. This hampers antibody validation and negatively impacts the reliability of western blot data, resulting worldwide in a considerable waste of reagents and labour. We argue that, to alleviate these problems there is a need to establish a database of reference MWs measured by SDS-PAGE. Using mass spectrometry as an orthogonal detection method, we acquired electrophoretic migration patterns for approximately 10'000 human proteins in five commonly used cell lines. We applied a robust internal calibration of migration to determine accurate and reproducible molecular weights. This in turn allows merging replicates to increase accuracy, but also enables comparing different cell lines. Mining of the data obtained highlights structural factors that affect migration of distinct classes of proteins. When combined with peptide coverage, the data produced recapitulates known post-translational modifications and differential splicing and can be used to formulate hypotheses on new or poorly known processing events. The full information is freely accessible as a web resource through a user friendly graphical interface (https://pumba.dcsr.unil.ch/). We anticipate that this database will be useful to investigators worldwide for troubleshooting western blot experiments, but could also contribute to the characterization of human proteoforms.


Asunto(s)
Bases de Datos de Proteínas , Electroforesis en Gel de Poliacrilamida , Proteínas , Humanos , Línea Celular , Espectrometría de Masas , Proteínas/química , Reproducibilidad de los Resultados , Peso Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA