Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros











Intervalo de año de publicación
1.
Nat Commun ; 15(1): 446, 2024 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-38199985

RESUMEN

Patients with corticosteroid-refractory acute graft-versus-host disease (aGVHD) have a low one-year survival rate. Identification and validation of novel targetable kinases in patients who experience corticosteroid-refractory-aGVHD may help improve outcomes. Kinase-specific proteomics of leukocytes from patients with corticosteroid-refractory-GVHD identified rho kinase type 1 (ROCK1) as the most significantly upregulated kinase. ROCK1/2 inhibition improved survival and histological GVHD severity in mice and was synergistic with JAK1/2 inhibition, without compromising graft-versus-leukemia-effects. ROCK1/2-inhibition in macrophages or dendritic cells prior to transfer reduced GVHD severity. Mechanistically, ROCK1/2 inhibition or ROCK1 knockdown interfered with CD80, CD86, MHC-II expression and IL-6, IL-1ß, iNOS and TNF production in myeloid cells. This was accompanied by impaired T cell activation by dendritic cells and inhibition of cytoskeletal rearrangements, thereby reducing macrophage and DC migration. NF-κB signaling was reduced in myeloid cells following ROCK1/2 inhibition. In conclusion, ROCK1/2 inhibition interferes with immune activation at multiple levels and reduces acute GVHD while maintaining GVL-effects, including in corticosteroid-refractory settings.


Asunto(s)
Enfermedad Injerto contra Huésped , Quinasas Asociadas a rho , Humanos , Animales , Ratones , Quinasas Asociadas a rho/genética , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Transducción de Señal , FN-kappa B , Corticoesteroides/farmacología , Corticoesteroides/uso terapéutico
2.
Biomolecules ; 13(3)2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36979484

RESUMEN

The actin nucleating and polymerizing formin-like 2 (FMNL2) is upregulated in several cancers and has been shown to play important roles in cell migration, invasion, cell-cell adhesion and filopodia formation. Here, using structured illumination microscopy we show that FMNL2 promotes rapid and highly dynamic filopodia formation in epithelial cells while remaining on the tip of the growing filopodia. This filopodia tip localization depends fully on its N-terminal myristoylation. We further show that FMNL2-dependent filopodia formation requires its serine 1072 phosphorylation within the diaphanous-autoregulatory domain (DAD) by protein kinase C (PKC) α. Consistent with this, filopodia formation depends on PKC activity and PKCα localizes to the base of growing filopodia. Thus, a PKCα-FMNL2 signaling module spatiotemporally controls dynamic filopodia formation.


Asunto(s)
Forminas , Proteína Quinasa C-alfa , Seudópodos , Actinas/metabolismo , Movimiento Celular , Fosforilación , Proteína Quinasa C-alfa/metabolismo , Seudópodos/metabolismo , Humanos , Forminas/metabolismo
3.
Nature ; 617(7961): 616-622, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36972684

RESUMEN

Steroid hormone receptors are ligand-binding transcription factors essential for mammalian physiology. The androgen receptor (AR) binds androgens mediating gene expression for sexual, somatic and behavioural functions, and is involved in various conditions including androgen insensitivity syndrome and prostate cancer1. Here we identified functional mutations in the formin and actin nucleator DAAM2 in patients with androgen insensitivity syndrome. DAAM2 was enriched in the nucleus, where its localization correlated with that of the AR to form actin-dependent transcriptional droplets in response to dihydrotestosterone. DAAM2 AR droplets ranged from 0.02 to 0.06 µm3 in size and associated with active RNA polymerase II. DAAM2 polymerized actin directly at the AR to promote droplet coalescence in a highly dynamic manner, and nuclear actin polymerization is required for prostate-specific antigen expression in cancer cells. Our data uncover signal-regulated nuclear actin assembly at a steroid hormone receptor necessary for transcription.


Asunto(s)
Actinas , Forminas , Proteínas Nucleares , Receptores Androgénicos , Transcripción Genética , Humanos , Actinas/metabolismo , Síndrome de Resistencia Androgénica/genética , Síndrome de Resistencia Androgénica/metabolismo , Andrógenos/farmacología , Andrógenos/metabolismo , Forminas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas Nucleares/metabolismo , Polimerizacion/efectos de los fármacos , Antígeno Prostático Específico/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptores Androgénicos/metabolismo , ARN Polimerasa II/metabolismo , Transducción de Señal/efectos de los fármacos , Esteroides/metabolismo , Esteroides/farmacología , Testosterona/análogos & derivados , Transcripción Genética/efectos de los fármacos
4.
Adv Sci (Weinh) ; 10(9): e2204896, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36691769

RESUMEN

Vesicle trafficking has emerged as an important process driving tumor progression through various mechanisms. Transforming growth factor beta (TGFß)-mediated secretion of Angiopoietin-like 4 (ANGPTL4) is important for cancer development. Here, Formin-like 2 (FMNL2) is identified to be necessary for ANGPTL4 trafficking and secretion in response to TGFß. Protein kinase C (PKC)-dependent phosphorylation of FMNL2 downstream of TGFß stimulation is required for cancer cell invasion as well as ANGPTL4 vesicle trafficking and secretion. Moreover, using super resolution microscopy, ANGPTL4 trafficking is actin-dependent with FMNL2 directly polymerizing actin at ANGPTL4-containing vesicles, which are associated with Rab8a and myosin Vb. This work uncovers a formin-controlled mechanism that transiently polymerizes actin directly at intracellular vesicles to facilitate their mobility. This mechanism may be important for the regulation of cancer cell metastasis and tumor progression.


Asunto(s)
Actinas , Factor de Crecimiento Transformador beta , Actinas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Forminas , Proteína 4 Similar a la Angiopoyetina
5.
Cells ; 11(22)2022 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-36429089

RESUMEN

Clostridioides bacteria are responsible for life threatening infections. Here, we show that in addition to actin, the binary toxins CDT, C2I, and Iota from Clostridioides difficile, botulinum, and perfrigens, respectively, ADP-ribosylate the actin-related protein Arp2 of Arp2/3 complex and its additional components ArpC1, ArpC2, and ArpC4/5. The Arp2/3 complex is composed of seven subunits and stimulates the formation of branched actin filament networks. This activity is inhibited after ADP-ribosylation of Arp2. Translocation of the ADP-ribosyltransferase component of CDT toxin into human colon carcinoma Caco2 cells led to ADP-ribosylation of cellular Arp2 and actin followed by a collapse of the lamellipodial extensions and F-actin network. Exposure of isolated mouse colon pieces to CDT toxin induced the dissolution of the enterocytes leading to luminal aggregation of cellular debris and the collapse of the mucosal organization. Thus, we identify the Arp2/3 complex as hitherto unknown target of clostridial ADP-ribosyltransferases.


Asunto(s)
Complejo 2-3 Proteico Relacionado con la Actina , Toxinas Bacterianas , Animales , Ratones , Humanos , Complejo 2-3 Proteico Relacionado con la Actina/metabolismo , Clostridioides , Actinas/metabolismo , Toxinas Bacterianas/farmacología , Toxinas Bacterianas/metabolismo , Células CACO-2 , ADP Ribosa Transferasas/farmacología , ADP Ribosa Transferasas/metabolismo , ADP-Ribosilación , Adenosina Difosfato/metabolismo
6.
Cell Rep ; 41(3): 111524, 2022 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-36260995

RESUMEN

The metabolic enzyme branched-chain amino acid transaminase 1 (BCAT1) drives cell proliferation in aggressive cancers such as glioblastoma. Here, we show that BCAT1 localizes to mitotic structures and has a non-metabolic function as a mitotic regulator. Furthermore, BCAT1 is required for chromosome segregation in cancer and induced pluripotent stem cells and tumor growth in human cerebral organoid and mouse syngraft models. Applying gene knockout and rescue strategies, we show that the BCAT1 CXXC redox motif is crucial for controlling cysteine sulfenylation specifically in mitotic cells, promoting Aurora kinase B localization to centromeres, and securing accurate chromosome segregation. These findings offer an explanation for the well-established role of BCAT1 in promoting cancer cell proliferation. In summary, our data establish BCAT1 as a component of the mitotic apparatus that safeguards mitotic fidelity through a moonlighting redox functionality.


Asunto(s)
Aminoácidos de Cadena Ramificada , Cisteína , Animales , Humanos , Ratones , Aurora Quinasa B , Modelos Animales de Enfermedad , Oxidación-Reducción , Transaminasas
7.
Development ; 148(1)2021 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-33199445

RESUMEN

Anterior mesoderm (AM) and definitive endoderm (DE) progenitors represent the earliest embryonic cell types that are specified during germ layer formation at the primitive streak (PS) of the mouse embryo. Genetic experiments indicate that both lineages segregate from Eomes-expressing progenitors in response to different Nodal signaling levels. However, the precise spatiotemporal pattern of the emergence of these cell types and molecular details of lineage segregation remain unexplored. We combined genetic fate labeling and imaging approaches with single-cell RNA sequencing (scRNA-seq) to follow the transcriptional identities and define lineage trajectories of Eomes-dependent cell types. Accordingly, all cells moving through the PS during the first day of gastrulation express Eomes AM and DE specification occurs before cells leave the PS from Eomes-positive progenitors in a distinct spatiotemporal pattern. ScRNA-seq analysis further suggested the immediate and complete separation of AM and DE lineages from Eomes-expressing cells as last common bipotential progenitor.


Asunto(s)
Linaje de la Célula , Endodermo/citología , Endodermo/metabolismo , Gastrulación , Mesodermo/citología , Mesodermo/metabolismo , Alelos , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/citología , Ratones , Modelos Biológicos , Línea Primitiva/embriología , Línea Primitiva/metabolismo , Células Madre/metabolismo , Proteínas de Dominio T Box/metabolismo , Factores de Tiempo , Transcripción Genética
8.
EMBO J ; 39(23): e107086, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33210291

RESUMEN

Maintenance of the mature blood cells requires controlled cell fate decisions by hematopoietic stem and progenitor cells (HSPCs). While our knowledge of the gene expression changes that facilitate differentiation has made a leap forward, less is known about the cellular triggers that induce them. Biedzinski et al (2020) now uncover a new intracellular mechanism that drives myeloid differentiation: Microtubule bundles squeeze the nucleus of HSPCs and form large invaginations, thus causing changes in chromatin organization. These microtubule-induced nuclear shape changes result in gene expression profiles that favor myeloid differentiation.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Diferenciación Celular , Constricción , Expresión Génica , Microtúbulos
9.
Nat Commun ; 9(1): 3524, 2018 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-30166526

RESUMEN

Anoikis is a form of apoptosis induced by cell detachment. Integrin inactivation plays a major role in the process but the exact signalling pathway is ill-defined. Here we identify an anoikis pathway using gliotoxin (GT), a virulence factor of the fungus Aspergillus fumigatus, which causes invasive aspergillosis in humans. GT prevents integrin binding to RGD-containing extracellular matrix components by covalently modifying cysteines in the binding pocket. As a consequence, focal adhesion kinase (FAK) is inhibited resulting in dephosphorylation of p190RhoGAP, allowing activation of RhoA. Sequential activation of ROCK, MKK4/MKK7 and JNK then triggers pro-apoptotic phosphorylation of Bim. Cells in suspension or lacking integrin surface expression are insensitive to GT but are sensitised to ROCK-MKK4/MKK7-JNK-dependent anoikis upon attachment to fibronectin or integrin upregulation. The same signalling pathway is triggered by FAK inhibition or inhibiting integrin αV/ß3 with Cilengitide. Thus, GT can target integrins to induce anoikis on lung epithelial cells.


Asunto(s)
Anoicis/fisiología , Gliotoxina/metabolismo , Transducción de Señal/fisiología , Factores de Virulencia/metabolismo , Amidas , Animales , Anoicis/genética , Línea Celular , Citometría de Flujo , Humanos , Immunoblotting , Inmunoprecipitación , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , MAP Quinasa Quinasa 7/genética , MAP Quinasa Quinasa 7/metabolismo , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Piridinas , Transducción de Señal/genética , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
10.
Oncotarget ; 8(44): 76686-76698, 2017 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-29100341

RESUMEN

Microtentacles are mostly microtubule-based cell protrusions that are formed by detached tumor cells. Here, we report that the formation of tumor cell microtentacles depends on the presence and dynamics of guanine nucleotide-binding proteins of the septin family, which are part of the cytoskeleton. In matrix-attached breast, lung, prostate and pancreas cancer cells, septins are associated with the cytosolic actin cytoskeleton. Detachment of cells causes redistribution of septins to the membrane, where microtentacle formation occurs. Forchlorfenuron, which inhibits septin functions, blocks microtentacle formation. The small GTPase Cdc42 and its effector proteins Borgs regulate septins and are essential for microtentacle formation. Dominant active and inactive Cdc42 inhibit microtentacle formation indicating that the free cycling of Cdc42 between its active and inactive state is essential for septin regulation and microtentacle formation. Cell attachment and aggregation models suggest that septins play an essential role in the metastatic behavior of tumor cells.

11.
Annu Rev Microbiol ; 71: 281-307, 2017 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-28657883

RESUMEN

Clostridium difficile is the cause of antibiotics-associated diarrhea and pseudomembranous colitis. The pathogen produces three protein toxins: C. difficile toxins A (TcdA) and B (TcdB), and C. difficile transferase toxin (CDT). The single-chain toxins TcdA and TcdB are the main virulence factors. They bind to cell membrane receptors and are internalized. The N-terminal glucosyltransferase and autoprotease domains of the toxins translocate from low-pH endosomes into the cytosol. After activation by inositol hexakisphosphate (InsP6), the autoprotease cleaves and releases the glucosyltransferase domain into the cytosol, where GTP-binding proteins of the Rho/Ras family are mono-O-glucosylated and, thereby, inactivated. Inactivation of Rho proteins disturbs the organization of the cytoskeleton and affects multiple Rho-dependent cellular processes, including loss of epithelial barrier functions, induction of apoptosis, and inflammation. CDT, the third C. difficile toxin, is a binary actin-ADP-ribosylating toxin that causes depolymerization of actin, thereby inducing formation of the microtubule-based protrusions. Recent progress in understanding of the toxins' actions include insights into the toxin structures, their interaction with host cells, and functional consequences of their actions.


Asunto(s)
ADP Ribosa Transferasas/toxicidad , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/toxicidad , Clostridioides difficile/metabolismo , Enterotoxinas/toxicidad , Células Epiteliales/efectos de los fármacos , Factores de Virulencia/toxicidad , ADP Ribosa Transferasas/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Toxinas Bacterianas/metabolismo , Citoesqueleto/efectos de los fármacos , Endocitosis , Enterotoxinas/metabolismo , Células Epiteliales/fisiología , Humanos , Microtúbulos/efectos de los fármacos , Factores de Virulencia/metabolismo
12.
Handb Exp Pharmacol ; 235: 179-206, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27316913

RESUMEN

Actin and the actin cytoskeleton play fundamental roles in host-pathogen interactions. Proper function of the actin cytoskeleton is crucial for innate and acquired immune defense. Bacterial toxins attack the actin cytoskeleton by targeting regulators of actin. Moreover, actin is directly modified by various bacterial protein toxins and effectors, which cause ADP-ribosylation or cross-linking of actin. Modification of actin can result in inhibition or stimulation of actin polymerization. Toxins, acting directly on actin, are reviewed.


Asunto(s)
Actinas/química , Adenosina Difosfato Ribosa/química , Toxinas Bacterianas/farmacología , Interacciones Huésped-Patógeno , Citoesqueleto de Actina/química , Animales , Reactivos de Enlaces Cruzados/química , Humanos , Polimerizacion
13.
Oncotarget ; 8(23): 37009-37022, 2017 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-27391068

RESUMEN

The lipolysis-stimulated lipoprotein receptor (LSR) is a lipoprotein receptor, serves as host receptor for clostridial iota-like toxins and is involved in the formation of tricellular contacts. Of particular interest is the role of LSR in progression of various cancers. Here we aimed to study the tumor growth of LSR-deficient colon carcinoma-derived cell lines HCT116 and CaCo-2 in a mouse xenograft model. Whereas knockout of LSR had no effect on tumor growth of HCT116 cells, we observed that CaCo-2 LSR knockout tumors grew to a smaller size than their wild-type counterparts. Histological analysis revealed increased apoptotic and necrotic cell death in a tumor originating from LSR-deficient CaCo-2 cells. LSR-deficient CaCo-2 cells exhibited increased cell proliferation in vitro and an altered epithelial morphology with impaired targeting of tricellulin to tricellular contacts. In addition, loss of LSR reduced the transepithelial electrical resistance of CaCo-2 cell monolayers and increased permeability for small molecules. Moreover, LSR-deficient CaCo-2 cells formed larger cysts in 3D culture than their wild-type counterparts. Our study provides evidence that LSR affects epithelial morphology and barrier formation in CaCo-2 cells and examines for the first time the effects of LSR deficiency on the tumor growth properties of colon carcinoma-derived cell lines.


Asunto(s)
Permeabilidad de la Membrana Celular , Neoplasias del Colon/metabolismo , Células Epiteliales/metabolismo , Receptores de Lipoproteína/metabolismo , Animales , Secuencia de Bases , Células CACO-2 , Proliferación Celular/genética , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Técnicas de Inactivación de Genes , Células HCT116 , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Noqueados , Ratones Desnudos , Receptores de Lipoproteína/genética , Trasplante Heterólogo , Carga Tumoral/genética
14.
Cell Microbiol ; 19(1)2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27341322

RESUMEN

Intoxication of eukaryotic cells by Photorhabdus luminescens toxin TccC3 induces cell rounding and detachment from the substratum within a few hours and compromises a number of cell functions like phagocytosis. Here, we used morphological and biochemical procedures to analyse the mechanism of TccC3 intoxication. Life imaging of TccC3-intoxicated HeLa cells transfected with AcGFP-actin shows condensation of F-actin into large aggregates. Life cell total internal reflection fluorescence (TIRF) microscopy of identically treated HeLa cells confirmed the formation of actin aggregates but also disassembly of F-actin stress fibres. Recombinant TccC3 toxin ADP-ribosylates purified skeletal and non-muscle actin at threonine148 leading to a strong propensity to polymerize and F-actin bundle formation as shown by TIRF and electron microscopy. Native gel electrophoresis shows strongly reduced binding of Thr148-ADP-ribosylated actin to the severing proteins gelsolin and its fragments G1 and G1-3, and to ADF/cofilin. Complexation of actin with these proteins inhibits its ADP-ribosylation. TIRF microscopy demonstrates rapid polymerization of Thr148-ADP-ribosylated actin to curled F-actin bundles even in the presence of thymosin ß4, gelsolin or G1-3. Thr148-ADP-ribosylated F-actin cannot be depolymerized by gelsolin or G1-3 as verified by TIRF, co-sedimentation and electron microscopy and shows reduced treadmilling as indicated by a lack of stimulation of its ATPase activity after addition of cofilin-1.


Asunto(s)
Actinas/metabolismo , Adenosina Difosfato/metabolismo , Toxinas Bacterianas/metabolismo , Photorhabdus/metabolismo , Agregación Patológica de Proteínas , Células Epiteliales/efectos de los fármacos , Células HeLa , Humanos , Microscopía Electrónica , Microscopía Fluorescente
15.
Nat Microbiol ; 1(8): 16108, 2016 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-27573114

RESUMEN

Clostridium difficile is the most common hospital acquired pathogen in the USA, and infection is, in many cases, fatal. Toxins A and B are its major virulence factors, but expression of a third toxin, known as C. difficile transferase (CDT), is increasingly common. An adenosine diphosphate (ADP)-ribosyltransferase that causes actin cytoskeletal disruption, CDT is typically produced by the major, hypervirulent strains and has been associated with more severe disease. Here, we show that CDT enhances the virulence of two PCR-ribotype 027 strains in mice. The toxin induces pathogenic host inflammation via a Toll-like receptor 2 (TLR2)-dependent pathway, resulting in the suppression of a protective host eosinophilic response. Finally, we show that restoration of TLR2-deficient eosinophils is sufficient for protection from a strain producing CDT. These findings offer an explanation for the enhanced virulence of CDT-expressing C. difficile and demonstrate a mechanism by which this binary toxin subverts the host immune response.


Asunto(s)
ADP Ribosa Transferasas/metabolismo , Proteínas Bacterianas/metabolismo , Clostridioides difficile/inmunología , Clostridioides difficile/patogenicidad , Infecciones por Clostridium/patología , Colon/inmunología , Eosinófilos/inmunología , Factores de Virulencia/metabolismo , Animales , Clostridioides difficile/clasificación , Clostridioides difficile/genética , Infecciones por Clostridium/microbiología , Modelos Animales de Enfermedad , Ratones , Ribotipificación
16.
Toxins (Basel) ; 8(4): 101, 2016 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-27043629

RESUMEN

The pathogenic bacteria Clostridium difficile, Clostridium perfringens and Clostridium botulinum produce the binary actin ADP-ribosylating toxins CDT, iota and C2, respectively. These toxins are composed of a transport component (B) and a separate enzyme component (A). When both components assemble on the surface of mammalian target cells, the B components mediate the entry of the A components via endosomes into the cytosol. Here, the A components ADP-ribosylate G-actin, resulting in depolymerization of F-actin, cell-rounding and eventually death. In the present study, we demonstrate that 4-bromobenzaldehyde N-(2,6-dimethylphenyl)semicarbazone (EGA), a compound that protects cells from multiple toxins and viruses, also protects different mammalian epithelial cells from all three binary actin ADP-ribosylating toxins. In contrast, EGA did not inhibit the intoxication of cells with Clostridium difficile toxins A and B, indicating a possible different entry route for this toxin. EGA does not affect either the binding of the C2 toxin to the cells surface or the enzyme activity of the A components of CDT, iota and C2, suggesting that this compound interferes with cellular uptake of the toxins. Moreover, for C2 toxin, we demonstrated that EGA inhibits the pH-dependent transport of the A component across cell membranes. EGA is not cytotoxic, and therefore, we propose it as a lead compound for the development of novel pharmacological inhibitors against clostridial binary actin ADP-ribosylating toxins.


Asunto(s)
ADP Ribosa Transferasas/toxicidad , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/toxicidad , Toxinas Botulínicas/toxicidad , Semicarbazonas/farmacología , Actinas/metabolismo , Adenosina Difosfato Ribosa/metabolismo , Animales , Membrana Celular/metabolismo , Chlorocebus aethiops , Células HeLa , Humanos , Transporte de Proteínas/efectos de los fármacos , Células Vero
17.
J Mol Biol ; 427(6 Pt A): 1224-38, 2015 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-25058685

RESUMEN

Clostridium botulinum C2 toxin, Clostridium perfringens iota toxin and Clostridium difficile CDT belong to the family of binary actin ADP-ribosylating toxins and are composed of a binding/translocation component and a separate enzyme component. The enzyme components ADP-ribosylate G-actin in the cytosol of target cells resulting in depolymerization of F-actin, cell rounding and cell death. The binding/translocation components bind to their cell receptors and form complexes with the respective enzyme components. After receptor-mediated endocytosis, the binding/translocation components form pores in membranes of acidified endosomes and the enzyme components translocate through these pores into the cytosol. This step is facilitated by the host cell chaperone heat shock protein 90 and peptidyl-prolyl cis/trans isomerases including cyclophilin A. Here, we demonstrate that a large isoform of cyclophilin A, the multi-domain enzyme cyclophilin 40 (Cyp40), binds to the enzyme components C2I, Ia and CDTa in vitro. Isothermal titration calorimetry revealed a direct binding to C2I with a calculated affinity of 101 nM and to Ia with an affinity of 1.01 µM. Closer investigation for the prototypic C2I revealed that binding to Cyp40 did not depend on its ADP-ribosyltransferase activity but was stronger for unfolded C2I. The interaction of C2I with Cyp40 was also demonstrated in lysates from C2-treated cells by pull-down. Treatment of cells with a non-immunosuppressive cyclosporine A derivative, which still binds to and inhibits the peptidyl-prolyl cis/trans isomerase activity of cyclophilins, protected cells from intoxication with C2, iota and CDT toxins, offering an attractive approach for development of novel therapeutic strategies against binary actin ADP-ribosylating toxins.


Asunto(s)
ADP Ribosa Transferasas/antagonistas & inhibidores , Proteínas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/antagonistas & inhibidores , Toxinas Botulínicas/antagonistas & inhibidores , Ciclofilinas/antagonistas & inhibidores , ADP Ribosa Transferasas/metabolismo , ADP Ribosa Transferasas/toxicidad , Actinas/metabolismo , Adenosina Difosfato/metabolismo , Animales , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/toxicidad , Toxinas Bacterianas/metabolismo , Toxinas Bacterianas/toxicidad , Toxinas Botulínicas/metabolismo , Toxinas Botulínicas/toxicidad , Chlorocebus aethiops , Ciclofilinas/metabolismo , Ciclosporina/farmacología , Células HeLa , Humanos , Transporte de Proteínas/efectos de los fármacos , Células Vero
18.
Cell Microbiol ; 17(4): 579-94, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25338512

RESUMEN

PTC3 and PTC5 are tripartite Tc (toxin complex) toxins from Photorhabdus luminescens, which consist of the binding component TcdA1, the linker component TcdB2 and the enzyme components TccC3 and TccC5 respectively. While PTC5 adenosine diphosphate (ADP)-ribosylates Rho proteins at Gln61/63 resulting in constitutive activation of the GTPases, PTC3 ADP-ribosylates actin at Thr148 thereby inducing actin polymerization. Here, we identified amino acids involved in ADP-ribosyltransferase activity of TccC3 and TccC5 and analysed the substrate specificity of Rho-activating TccC5. We compared the time dependency of Rho protein activation by PTC5 in HeLa cells with the effects of Escherichia coli cytotoxic necrotizing factor 1, which activates Rho GTPases by deamidation of Gln61/63. Using a luciferase reporter assay, we show that PTC5 and PTC3 stimulated gene transcription via myocardin-related transcription factor A (also called MAL) and AP1. MAL activation by PTC5 involved Rho kinase and formins. Activation of AP1 by PTC5 occurred via two MAP kinase pathways involving extracellular signal-regulated kinase and Jun kinase respectively.


Asunto(s)
ADP Ribosa Transferasas/metabolismo , Actinas/metabolismo , Toxinas Bacterianas/metabolismo , Photorhabdus/enzimología , Transactivadores/metabolismo , Factor de Transcripción AP-1/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Genes Reporteros , Células HeLa , Humanos , Luciferasas/análisis , Luciferasas/genética , Procesamiento Proteico-Postraduccional , Transcripción Genética
19.
Toxins (Basel) ; 5(11): 2241-57, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24284827

RESUMEN

Prostate cancer is the most common malignancy, accounting for about 25% of all incident cases among men in industrialized countries. The human androgen-dependent prostate cancer cell line LNCaP, which is derived from a metastatic lesion of human prostatic adenocarcinoma, is frequently used to study prostate cancer associated signaling pathways in vitro. Recently it was described that Rho GTPase activation in these cells leads to apoptotic responses. We used the bacterial toxins CNFy and CNF1, which specifically and directly activate Rho GTPases by deamidation of a single glutamine. We asked whether these Rho activators could induce apoptosis in LNCaP cells. Our results indicate that RhoA activation, induced by CNFy, does lead to intrinsic apoptosis of the cells. Analysis of the underlying signaling pathway reveals that apoptosis induction requires the activity of Rho kinase (ROCK) and myosin activation, an apoptotic pathway previously identified in cancer stem cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Toxinas Bacterianas/farmacología , Proteínas de Escherichia coli/farmacología , Yersinia , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Humanos , Masculino , Miosinas/genética , Miosinas/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Recombinantes/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rho/genética , Proteína de Unión al GTP rhoA/genética
20.
J Biol Chem ; 287(30): 24929-40, 2012 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-22665487

RESUMEN

TpeL is a member of the family of clostridial glucosylating toxins produced by Clostridium perfringens type A, B, and C strains. In contrast to other members of this toxin family, it lacks a C-terminal polypeptide repeat domain, which is suggested to be involved in target cell binding. It was shown that the glucosyltransferase domain of TpeL modifies Ras in vitro by mono-O-glucosylation or mono-O-GlcNAcylation (Nagahama, M., Ohkubo, A., Oda, M., Kobayashi, K., Amimoto, K., Miyamoto, K., and Sakurai, J. (2011) Infect. Immun. 79, 905-910). Here we show that TpeL preferably utilizes UDP-N-acetylglucosamine (UDP-GlcNAc) as a sugar donor. Change of alanine 383 of TpeL to isoleucine turns the sugar donor preference from UDP-GlcNAc to UDP-glucose. In contrast to previous studies, we show that Rac is a poor substrate in vitro and in vivo and requires 1-2 magnitudes higher toxin concentrations for modification by TpeL. The toxin is autoproteolytically processed in the presence of inositol hexakisphosphate (InsP(6)) by an intrinsic cysteine protease domain, located next to the glucosyltransferase domain. A C-terminally extended TpeL full-length variant (TpeL1-1779) induces apoptosis in HeLa cells (most likely by mono-O-GlcNAcylation of Ras), and inhibits Ras signaling including Ras-Raf interaction and ERK activation. In addition, TpeL blocks Ras signaling in rat pheochromocytoma PC12 cells. TpeL is a glucosylating toxin, which modifies Ras and induces apoptosis in target cells without having a typical C-terminal polypeptide repeat domain.


Asunto(s)
Acetilglucosamina/metabolismo , Toxinas Bacterianas/metabolismo , Clostridium perfringens/enzimología , Glicosiltransferasas/metabolismo , Proteína Oncogénica p21(ras)/metabolismo , Proteolisis , Acetilglucosamina/genética , Animales , Apoptosis/genética , Toxinas Bacterianas/genética , Clostridium perfringens/genética , Activación Enzimática/genética , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Glicosilación , Glicosiltransferasas/genética , Células HeLa , Humanos , Proteína Oncogénica p21(ras)/genética , Células PC12 , Ratas , Azúcares de Uridina Difosfato/genética , Azúcares de Uridina Difosfato/metabolismo , Quinasas raf/genética , Quinasas raf/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA