Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 80(9): 258, 2023 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-37594630

RESUMEN

HtrA2/Omi is a mitochondrial serine protease with ascribed pro-apoptotic as well as pro-necroptotic functions. Here, we establish that HtrA2/Omi also controls parthanatos, a third modality of regulated cell death. Deletion of HtrA2/Omi protects cells from parthanatos while reconstitution with the protease restores the parthanatic death response. The effects of HtrA2/Omi on parthanatos are specific and cannot be recapitulated by manipulating other mitochondrial proteases such as PARL, LONP1 or PMPCA. HtrA2/Omi controls parthanatos in a manner mechanistically distinct from its action in apoptosis or necroptosis, i.e., not by cleaving cytosolic IAP proteins but rather exerting its effects without exiting mitochondria, and downstream of PARP-1, the first component of the parthanatic signaling cascade. Also, previously identified or candidate substrates of HtrA2/Omi such as PDXDC1, VPS4B or moesin are not cleaved and dispensable for parthanatos, whereas DBC-1 and stathmin are cleaved, and thus represent potential parthanatic downstream mediators of HtrA2/Omi. Moreover, mass-spectrometric screening for novel parthanatic substrates of HtrA2/Omi revealed that the induction of parthanatos does not cause a substantial proteolytic cleavage or major alterations in the abundance of mitochondrial proteins. Resolving these findings, reconstitution of HtrA2/Omi-deficient cells with a catalytically inactive HtrA2/Omi mutant restored their sensitivity against parthanatos to the same level as the protease-active HtrA2/Omi protein. Additionally, an inhibitor of HtrA2/Omi's protease activity did not confer protection against parthanatic cell death. Our results demonstrate that HtrA2/Omi controls parthanatos in a protease-independent manner, likely via novel, unanticipated functions as a scaffolding protein and an interaction with so far unknown mitochondrial proteins.


Asunto(s)
Parthanatos , Serina Proteasas/genética , Necroptosis , Serina Endopeptidasas/genética , Proteínas Mitocondriales/genética
2.
Infection ; 51(3): 561-565, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37014548

RESUMEN

Theodor Escherich (1857-1911) was one of the key players in early paediatric infectious diseases (PID). In fact, he can be regarded as the first paediatric infectious diseases physician and the founder of this subspecialty. During his long years in service for children, he spent 6 years at the Dr von Hauner children's hospital (1884-1890), laying the foundations for PID clinical care and research in Munich. Walter Marget, founder of this journal and co-founder of the German Society for Infectious Diseases (DGI) graduated from medical school in 1946 and practised in Munich since 1967. His tireless efforts went into establishing close links between clinical paediatrics and microbiological diagnostics culminating in the foundation of the Department of Antimicrobial Therapy and Infection Epidemiology at the Dr von Hauner children's hospital. Walter Marget was a key figure for PID in Germany having trained and supported many clinician scientists who followed in his footsteps. This article gives a brief overview of the history of PID in Munich while commemorating Walter Marget and his achievements in this field and for INFECTION.


Asunto(s)
Enfermedades Transmisibles , Dermatitis , Masculino , Humanos , Niño , Historia del Siglo XX , Alemania , Infectología
3.
Eur Respir J ; 60(4)2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35450969

RESUMEN

BACKGROUND: Prostaglandin E2 (PGE2) increases pulmonary vascular permeability by activation of the PGE2 receptor 3 (EP3), which may explain adverse pulmonary effects of the EP1/EP3 receptor agonist sulprostone in patients. In addition, PGE2 contributes to pulmonary oedema in response to platelet-activating factor (PAF). PAF increases endothelial permeability by recruiting the cation channel transient receptor potential canonical 6 (TRPC6) to endothelial caveolae via acid sphingomyelinase (ASMase). Yet, the roles of PGE2 and EP3 in this pathway are unknown. We hypothesised that EP3 receptor activation may increase pulmonary vascular permeability by activation of TRPC6, and thus, synergise with ASMase-mediated TRPC6 recruitment in PAF-induced lung oedema. METHODS: In isolated lungs, we measured increases in endothelial calcium (ΔCa2+) or lung weight (Δweight), and endothelial caveolar TRPC6 abundance as well as phosphorylation. RESULTS: PAF-induced ΔCa2+ and Δweight were attenuated in EP3-deficient mice. Sulprostone replicated PAF-induced ΔCa2+ and Δweight which were blocked by pharmacological/genetic inhibition of TRPC6, ASMase or Src-family kinases (SrcFK). PAF, but not sulprostone, increased TRPC6 abundance in endothelial caveolae. Immunoprecipitation revealed PAF- and sulprostone-induced tyrosine-phosphorylation of TRPC6 that was prevented by inhibition of phospholipase C (PLC) or SrcFK. PLC inhibition also blocked sulprostone-induced ΔCa2+ and Δweight, as did inhibition of SrcFK or inhibitory G-protein (Gi) signalling. CONCLUSIONS: EP3 activation triggers pulmonary oedema via Gi-dependent activation of PLC and subsequent SrcFK-dependent tyrosine phosphorylation of TRPC6. In PAF-induced lung oedema, this TRPC6 activation coincides with ASMase-dependent caveolar recruitment of TRPC6, resulting in rapid endothelial Ca2+ influx and barrier failure.


Asunto(s)
Edema Pulmonar , Animales , Calcio/metabolismo , Edema , Células Endoteliales/metabolismo , Proteínas de Unión al GTP/metabolismo , Pulmón/metabolismo , Ratones , Factor de Activación Plaquetaria , Esfingomielina Fosfodiesterasa , Canal Catiónico TRPC6 , Fosfolipasas de Tipo C/metabolismo , Tirosina , Familia-src Quinasas
4.
Eur J Immunol ; 49(10): 1457-1973, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31633216

RESUMEN

These guidelines are a consensus work of a considerable number of members of the immunology and flow cytometry community. They provide the theory and key practical aspects of flow cytometry enabling immunologists to avoid the common errors that often undermine immunological data. Notably, there are comprehensive sections of all major immune cell types with helpful Tables detailing phenotypes in murine and human cells. The latest flow cytometry techniques and applications are also described, featuring examples of the data that can be generated and, importantly, how the data can be analysed. Furthermore, there are sections detailing tips, tricks and pitfalls to avoid, all written and peer-reviewed by leading experts in the field, making this an essential research companion.


Asunto(s)
Alergia e Inmunología/normas , Separación Celular/métodos , Separación Celular/normas , Citometría de Flujo/métodos , Citometría de Flujo/normas , Consenso , Humanos , Fenotipo
5.
Mediators Inflamm ; 2020: 3650508, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32410851

RESUMEN

Tumor necrosis factor (TNF) is a well-known mediator of sepsis. In many cases, sepsis results in multiple organ injury including the lung with acute respiratory distress syndrome (ARDS). More than 20-year-old studies have suggested that TNF may be directly responsible for organ injury during sepsis. However, these old studies are inconclusive, because they relied on human rather than conspecific TNF, which was contaminated with endotoxin in most studies. In this study, we characterized the direct effects of intravenous murine endotoxin-free TNF on cardiovascular functions and organ injury in mice with a particular focus on the lungs. Because of the relevance of the acid sphingomyelinase in sepsis, ARDS, and caspase-independent cell death, we also included acid sphingomyelinase-deficient (ASM-/-) mice. ASM-/- and wild-type (WT) mice received 50 µg endotoxin-free murine TNF intravenously alone or in combination with the pan-caspase inhibitor carbobenzoxy-valyl-alanyl-aspartyl-[O-methyl]-fluoromethylketone (zVAD) and were ventilated at low tidal volume while lung mechanics were followed. Blood pressure was stabilized by intra-arterial fluid support, and body temperature was kept at 37°C to delay lethal shock and to allow investigation of blood gases, lung histopathology, proinflammatory mediators, and microvascular permeability 6 hours after TNF application. Besides the lungs, also the kidneys and liver were examined. TNF elicited the release of inflammatory mediators and a high mortality rate, but failed to injure the lungs, kidneys, or liver of healthy mice significantly within 6 hours. Mortality in WT mice was most likely due to sepsis-like shock, as indicated by metabolic acidosis, high procalcitonin levels, and cardiovascular failure. ASM-/- mice were protected from TNF-induced hypotension and reflex tachycardia and also from mortality. In WT mice, intravenous exogenous TNF does not cause organ injury but induces a systemic inflammatory response with cardiovascular failure, in which the ASM plays a role.


Asunto(s)
Lesión Pulmonar/metabolismo , Choque/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Animales , Permeabilidad Capilar , Endotoxinas/metabolismo , Femenino , Inflamación , Mediadores de Inflamación/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Pulmón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microcirculación , Neutrófilos/metabolismo , Oligopéptidos/farmacología , Permeabilidad , Respiración Artificial , Sepsis
6.
Cell Commun Signal ; 17(1): 90, 2019 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-31382980

RESUMEN

BACKGROUND: Binding of tumor necrosis factor (TNF) to TNF-receptor 1 (TNF-R1) can induce either cell survival or cell death. The selection between these diametrically opposed effects depends on the subcellular location of TNF-R1: plasma membrane retention leads to survival, while endocytosis leads to cell death. How the respective TNF-R1 associated signaling complexes are recruited to the distinct subcellular location is not known. Here, we identify palmitoylation of TNF-R1 as a molecular mechanism to achieve signal diversification. METHODS: Human monocytic U937 cells were analyzed. Palmitoylated proteins were enriched by acyl resin assisted capture (AcylRAC) and analyzed by western blot and mass spectrometry. Palmitoylation of TNF-R1 was validated by metabolic labeling. TNF induced depalmitoylation and involvement of APT2 was analyzed by enzyme activity assays, pharmacological inhibition and shRNA mediated knock-down. TNF-R1 palmitoylation site analysis was done by mutated TNF-R1 expression in TNF-R1 knock-out cells. Apoptosis (nuclear DNA fragmentation, caspase 3 assays), NF-κB activation and TNF-R1 internalization were used as biological readouts. RESULTS: We identify dynamic S-palmitoylation as a new mechanism that controls selective TNF signaling. TNF-R1 itself is constitutively palmitoylated and depalmitoylated upon ligand binding. We identified the palmitoyl thioesterase APT2 to be involved in TNF-R1 depalmitoylation and TNF induced NF-κB activation. Mutation of the putative palmitoylation site C248 interferes with TNF-R1 localization to the plasma membrane and thus, proper signal transduction. CONCLUSIONS: Our results introduce palmitoylation as a new layer of dynamic regulation of TNF-R1 induced signal transduction at a very early step of the TNF induced signaling cascade. Understanding the underlying mechanism may allow novel therapeutic options for disease treatment in future.


Asunto(s)
Lipoilación , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal , Línea Celular , Regulación de la Expresión Génica , Humanos , FN-kappa B/metabolismo , Transporte de Proteínas , Tioléster Hidrolasas/metabolismo
7.
Biochim Biophys Acta Mol Cell Res ; 1864(11 Pt B): 2147-2161, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28577894

RESUMEN

Proteases control most of the physiological processes that occur in a cell. This particularly applies to apoptosis, the most well-studied form of cell death, where proteolysis by cysteine-aspartic proteases (caspases) is the primary mechanism for both initiation and execution of cell suicide. In contrast, the impact of proteolysis on other, non-apoptotic cell death pathways (summarized under the term "regulated necrosis", RN) has long been enigmatic, but has clearly been confirmed by a number of recent groundbreaking discoveries. Here, we review these discoveries and provide an overview on the role of proteolysis in known forms of RN, with a particular focus on necroptosis and pyroptosis, and their regulation by deubiquitinases, apoptotic and inflammatory caspases. This article is part of a Special Issue entitled: Proteolysis as a Regulatory Event in Pathophysiology edited by Stefan Rose-John.


Asunto(s)
Apoptosis/genética , Necrosis/genética , Proteolisis , Piroptosis/genética , Caspasas/genética , Enzimas Desubicuitinizantes/genética , Humanos , Péptido Hidrolasas/genética , Transducción de Señal
8.
Cell Mol Life Sci ; 73(11-12): 2183-93, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27048810

RESUMEN

Regulated cell death is one major factor to ensure homoeostasis in multicellular organisms. For decades, apoptosis was considered as the sole form of regulated cell death, whereas necrosis was believed to be accidental and unregulated. Due to this view, research on necrosis was somewhat neglected, especially in the field of anti-cancer treatment. However, new interest in necrosis has been sparked by the recent discovery of different forms of necrosis that show indeed regulated pathways. More and more studies now address the molecular pathways of regulated necrosis and its connections within the cellular signaling networks. Necroptosis, a subform of regulated necrosis, has so far hardly been focused on with regard to a future treatment of cancer patients and may emerge as a novel and effective approach to eliminate tumor cells. However, and similar to apoptosis, tumor cells can develop resistances against necroptosis to ensure their own survival. In this context, new molecules that enhance necroptosis are currently being identified to overcome such resistances. This review discusses cancer and necroptosis as friends or foes, i.e. the options to exploit necroptosis in anti-cancer therapies ("foes"), but also potential limitations that may block or actually cause necroptosis to act in a protumoral manner ("friends"). The balance between these two possible roles will determine whether necroptosis can indeed be used as a promising tool for early diagnosis of tumors, prevention of metastasis and anti-cancer treatment.


Asunto(s)
Apoptosis/fisiología , Necrosis/patología , Neoplasias/patología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Humanos , Neoplasias/diagnóstico , Neoplasias/terapia , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
9.
Gastroenterology ; 146(1): 278-90, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24120475

RESUMEN

BACKGROUND & AIMS: Tumor necrosis factor-related apoptosis inducing ligand (TRAIL-R1) (TNFRSF10A) and TRAIL-R2 (TNFRSF10B) on the plasma membrane bind ligands that activate apoptotic and other signaling pathways. Cancer cells also might have TRAIL-R2 in the cytoplasm or nucleus, although little is known about its activities in these locations. We investigated the functions of nuclear TRAIL-R2 in cancer cell lines. METHODS: Proteins that interact with TRAIL-R2 initially were identified in pancreatic cancer cells by immunoprecipitation, mass spectrometry, and immunofluorescence analyses. Findings were validated in colon, renal, lung, and breast cancer cells. Functions of TRAIL-R2 were determined from small interfering RNA knockdown, real-time polymerase chain reaction, Drosha-activity, microRNA array, proliferation, differentiation, and immunoblot experiments. We assessed the effects of TRAIL-R2 overexpression or knockdown in human pancreatic ductal adenocarcinoma (PDAC) cells and their ability to form tumors in mice. We also analyzed levels of TRAIL-R2 in sections of PDACs and non-neoplastic peritumoral ducts from patients. RESULTS: TRAIL-R2 was found to interact with the core microprocessor components Drosha and DGCR8 and the associated regulatory proteins p68, hnRNPA1, NF45, and NF90 in nuclei of PDAC and other tumor cells. Knockdown of TRAIL-R2 increased Drosha-mediated processing of the let-7 microRNA precursor primary let-7 (resulting in increased levels of mature let-7), reduced levels of the let-7 targets (LIN28B and HMGA2), and inhibited cell proliferation. PDAC tissues from patients had higher levels of nuclear TRAIL-R2 than non-neoplastic pancreatic tissue, which correlated with increased nuclear levels of HMGA2 and poor outcomes. Knockdown of TRAIL-R2 in PDAC cells slowed their growth as orthotopic tumors in mice. Reduced nuclear levels of TRAIL-R2 in cultured pancreatic epithelial cells promoted their differentiation. CONCLUSIONS: Nuclear TRAIL-R2 inhibits maturation of the microRNA let-7 in pancreatic cancer cell lines and increases their proliferation. Pancreatic tumor samples have increased levels of nuclear TRAIL-R2, which correlate with poor outcome of patients. These findings indicate that in the nucleus, death receptors can function as tumor promoters and might be therapeutic targets.


Asunto(s)
Apoptosis/fisiología , Carcinoma Ductal Pancreático/metabolismo , MicroARNs/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular , Neoplasias del Colon/metabolismo , Humanos , Neoplasias Renales/metabolismo , Neoplasias Pulmonares/metabolismo , Ratones , Ratones SCID , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/fisiología
10.
Cell Commun Signal ; 13: 25, 2015 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-25925126

RESUMEN

BACKGROUND: One hallmark of cancer cells is their ability to evade physiologic signals causing regulated cell death (RCD). Correspondingly, TRAIL-based therapies to eliminate human cancer cells via enforced induction of apoptosis have been established and represent a promising approach in anti-cancer research. However, due to frequently appearing intrinsic or acquired resistances of tumor cells against apoptosis, TRAIL-based apoptotic strategies for the treatment of cancer patients have shown limited efficacy. As a potential alternative, regulated necrosis (and necroptosis triggered e.g. by TRAIL receptors 1/2) has recently gained considerable attention. Regulated necrosis represents a mode of RCD molecularly distinct from apoptosis whose potential in anti-cancer therapy is almost uncharacterized. Since in most cancer cells survival pathways counteract the effects of TRAIL-induced RCD, sensitizers such as cycloheximide (CHX) are frequently added in cell culture to overcome this problem. Unfortunately, those sensitizers are cytotoxic and therefore not suitable for the treatment of cancer patients. Here, we have alternatively employed homoharringtonine (HHT), a plant alkaloid which was recently approved by the U. S. Food and Drug Administration to treat patients with chronic myeloid lymphoma. RESULTS: We show that HHT is an efficient sensitizer for TRAIL-induced necroptosis in multiple human cancer cell lines. In addition, HHT-enhanced TRAIL-mediated necroptosis occurs via the same signaling pathways (involving RIPK1/RIPK3/MLKL) as CHX-enhanced necroptosis. Importantly, consecutive treatment schedules of necroptosis and apoptosis in either combination revealed remarkable additive effects not reached by repetitive apoptotic treatments alone. CONCLUSIONS: Taken together, our data demonstrate that HHT can replace harmful substances such as CHX to sensitize human cancer cells to TRAIL-induced necroptosis. Thus, HHT represents a promising enhancer in TRAIL-based necroptotic anti-cancer therapies also in patients.


Asunto(s)
Antineoplásicos/farmacología , Harringtoninas/farmacología , Neoplasias/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Línea Celular Tumoral , Cicloheximida/farmacología , Homoharringtonina , Humanos , Necrosis , Neoplasias/metabolismo , Neoplasias/patología , Inhibidores de la Síntesis de la Proteína/farmacología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
11.
Eur J Pediatr ; 174(3): 355-64, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25169065

RESUMEN

UNLABELLED: Otitis media is a common pediatric disease and the main reason for antibiotic prescription in children. Before implementation of routine childhood pneumococcal vaccination in Germany, serotypes contained in the seven-valent pneumococcal conjugate vaccine (PCV) were among the most frequent pneumococcal serotypes responsible for acute otitis media (AOM). This report describes the first 3 years of a prospective, multicenter, epidemiological cross-sectional study examining the bacteriology of middle ear fluids (MEF) and nasopharyngeal swabs (NPS) of children 2 months to 5 years of age with spontaneously perforated AOM in the era of routine pneumococcal vaccination. MEF was obtained from 963 subjects; NPS from 877. Reported case numbers steeply decreased over the three study years even though the recruiting base remained the same. Among subjects with relevant bacterial growth in their MEF swabs, 113 (11.7%) had Streptococcus pyogenes, 97 (10.1%) Staphylococcus aureus, 88 (9.1%) Streptococcus pneumoniae, 63 (6.5%) Haemophilus influenzae, and 8 (0.8%) Moraxella catarrhalis. S. pneumoniae isolates decreased from 41 (9.3%) in year 1 to 12 (5.7%) in year 3 (p = 0.128). PCV7 serotypes accounted for only 7.9% (n = 7) of isolated pneumococci. Of the 877 subjects with NPS cultures, 465 (53.0%) carried S. pneumoniae, 314 (35.8%) H. influenzae, 292 (33.3%) M. catarrhalis, and 110 (12.5%) S. pyogenes; 79.4% (n = 765) of the children were vaccinated with at least one dose of PCV. Carriage of pneumococci was slightly lower in vaccinated (47.8%) than in unvaccinated (52.7%) children (p = 0.254). PCV7 serotypes were carried by 9.6% of unvaccinated children but by only 4.2% of vaccinated children, resulting in a 56.3% vaccine effectiveness. CONCLUSIONS: Following universal PCV7 immunization, a clear epidemiological impact of pneumococcal conjugate vaccination was observed as PCV7 serotypes have almost disappeared among AOM.


Asunto(s)
Otitis Media/microbiología , Vacunas Neumococicas/inmunología , Perforación de la Membrana Timpánica/microbiología , Vacunas Conjugadas/inmunología , Enfermedad Aguda , Preescolar , Estudios Transversales , Oído Medio/microbiología , Femenino , Alemania/epidemiología , Haemophilus influenzae/aislamiento & purificación , Humanos , Lactante , Masculino , Moraxella catarrhalis/aislamiento & purificación , Nasofaringe/microbiología , Otitis Media/epidemiología , Otitis Media/inmunología , Vacunas Neumococicas/sangre , Estudios Prospectivos , Serogrupo , Staphylococcus aureus/aislamiento & purificación , Streptococcus pneumoniae/aislamiento & purificación , Streptococcus pyogenes/aislamiento & purificación
12.
Cell Mol Life Sci ; 71(2): 331-48, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23760205

RESUMEN

Programmed necrosis is important in many (patho)physiological settings. For specific therapeutic intervention, however, a better knowledge is required whether necrosis occurs through one single "core program" or through several independent pathways. Previously, the poly(ADP-ribose) polymerase (PARP) pathway has been suggested as a crucial element of tumor necrosis factor (TNF)-mediated necroptosis. Here, we show that TNF-induced necroptosis and the PARP pathway represent distinct and independent routes to programmed necrosis. First, DNA-alkylating agents such as 1-methyl-3-nitro-1-nitrosoguanidine (MNNG) or methyl methanesulfonate rapidly activate the PARP pathway, whereas this is a late and secondary event in TNF-induced necroptosis. Second, inhibition of the PARP pathway does not protect against TNF-induced necroptosis, e.g., the PARP-1 inhibitor 3-AB prevented MNNG- but not TNF-induced adenosine-5'-triposphate depletion, translocation of apoptosis-inducing factor, and necrosis. Likewise, olaparib, a more potent and selective PARP-1 inhibitor failed to block TNF-induced necroptosis, identical to knockdown/knockout of PARP-1, pharmacologic and genetic interference with c-Jun N-terminal kinases and calpain/cathepsin proteases as further components of the PARP pathway. Third, interruption of TNF-induced necroptosis by interference with ceramide generation, RIP1 or RIP3 function or by the radical scavenger butylated hydroxyanisole did not prevent programmed necrosis through the PARP pathway. In summary, our results suggest that the currently established role of the PARP pathway in TNF-induced necroptosis needs to be revised, with consequences for the design of future therapeutic strategies.


Asunto(s)
Apoptosis/efectos de los fármacos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Animales , Antineoplásicos Alquilantes/farmacología , Benzamidas/farmacología , Calpaína/metabolismo , Catepsinas/metabolismo , Línea Celular , Ceramidas/metabolismo , Depuradores de Radicales Libres/farmacología , Guanidinas/farmacología , Células HEK293 , Células HT29 , Células HeLa , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Células Jurkat , Células MCF-7 , Metilmetanosulfonato/farmacología , Ratones , Necrosis , Proteínas de Complejo Poro Nuclear/metabolismo , Ftalazinas/farmacología , Piperazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/genética , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
13.
Mar Drugs ; 13(7): 4470-91, 2015 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-26204945

RESUMEN

Pancreatic cancer is one of the most aggressive cancer entities, with an extremely poor 5-year survival rate. Therefore, novel therapeutic agents with specific modes of action are urgently needed. Marine organisms represent a promising source to identify new pharmacologically active substances. Secondary metabolites derived from marine algae are of particular interest. The present work describes cellular and molecular mechanisms induced by an HPLC-fractionated, hydrophilic extract derived from the Baltic brown seaweed Fucus vesiculosus (Fv1). Treatment with Fv1 resulted in a strong inhibition of viability in various pancreatic cancer cell lines. This extract inhibited the cell cycle of proliferating cells due to the up-regulation of cell cycle inhibitors, shown on the mRNA (microarray data) and protein level. As a result, cells were dying in a caspase-independent manner. Experiments with non-dividing cells showed that proliferation is a prerequisite for the effectiveness of Fv1. Importantly, Fv1 showed low cytotoxic activity against non-malignant resting T cells and terminally differentiated cells like erythrocytes. Interestingly, accelerated killing effects were observed in combination with inhibitors of autophagy. Our in vitro data suggest that Fv1 may represent a promising new agent that deserves further development towards clinical application.


Asunto(s)
Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Fucus/química , Neoplasias Pancreáticas/tratamiento farmacológico , Extractos Vegetales/farmacología , Autofagia/efectos de los fármacos , Caspasas/fisiología , Línea Celular Tumoral , Humanos
14.
Traffic ; 13(1): 108-19, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21985295

RESUMEN

Chediak-Higashi syndrome is an autosomal recessive disorder that affects vesicle morphology. The Chs1/Lyst protein is a member of the BEige And CHediak family of proteins. The absence of Chs1/Lyst gives rise to enlarged lysosomes. Lysosome size is regulated by a balance between vesicle fusion and fission and can be reversibly altered by acidifying the cytoplasm using Acetate Ringer's or by incubating with the drug vacuolin-1. We took advantage of these procedures to determine rates of lysosome fusion and fission in the presence or absence of Chs1/Lyst. Here, we show by microscopy, flow cytometry and in vitro fusion that the absence of the Chs1/Lyst protein does not increase the rate of lysosome fusion. Rather, our data indicate that loss of this protein decreases the rate of lysosome fission. We further show that overexpression of the Chs1/Lyst protein gives rise to a faster rate of lysosome fission. These results indicate that Chs1/Lyst regulates lysosome size by affecting fission.


Asunto(s)
Síndrome de Chediak-Higashi , Lisosomas/ultraestructura , Macrófagos/ultraestructura , Proteínas/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Animales , Células Cultivadas , Síndrome de Chediak-Higashi/metabolismo , Síndrome de Chediak-Higashi/patología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Citometría de Flujo , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas/genética , Proteínas de Transporte Vesicular/genética
15.
BMC Cancer ; 14: 74, 2014 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-24507727

RESUMEN

BACKGROUND: The cytokine TRAIL represents one of the most promising candidates for the apoptotic elimination of tumor cells, either alone or in combination therapies. However, its efficacy is often limited by intrinsic or acquired resistance of tumor cells to apoptosis. Programmed necrosis is an alternative, molecularly distinct mode of programmed cell death that is elicited by TRAIL under conditions when the classical apoptosis machinery fails or is actively inhibited. The potential of TRAIL-induced programmed necrosis in tumor therapy is, however, almost completely uncharacterized. We therefore investigated its impact on a panel of tumor cell lines of wide-ranging origin. METHODS: Cell death/viability was measured by flow cytometry/determination of intracellular ATP levels/crystal violet staining. Cell surface expression of TRAIL receptors was detected by flow cytometry, expression of proteins by Western blot. Ceramide levels were quantified by high-performance thin layer chromatography and densitometric analysis, clonogenic survival of cells was determined by crystal violet staining or by soft agarose cloning. RESULTS: TRAIL-induced programmed necrosis killed eight out of 14 tumor cell lines. Clonogenic survival was reduced in all sensitive and even one resistant cell lines tested. TRAIL synergized with chemotherapeutics in killing tumor cell lines by programmed necrosis, enhancing their effect in eight out of 10 tested tumor cell lines and in 41 out of 80 chemotherapeutic/TRAIL combinations. Susceptibility/resistance of the investigated tumor cell lines to programmed necrosis seems to primarily depend on expression of the pro-necrotic kinase RIPK3 rather than the related kinase RIPK1 or cell surface expression of TRAIL receptors. Furthermore, interference with production of the lipid ceramide protected all tested tumor cell lines. CONCLUSIONS: Our study provides evidence that TRAIL-induced programmed necrosis represents a feasible approach for the elimination of tumor cells, and that this treatment may represent a promising new option for the future development of combination therapies. Our data also suggest that RIPK3 expression may serve as a potential predictive marker for the sensitivity of tumor cells to programmed necrosis and extend the previously established role of ceramide as a key mediator of death receptor-induced programmed necrosis (and thus as a potential target for future therapies) also to the tumor cell lines examined here.


Asunto(s)
Apoptosis/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/envenenamiento , Antineoplásicos/envenenamiento , Western Blotting , Muerte Celular/genética , Citometría de Flujo/métodos , Regulación Neoplásica de la Expresión Génica , Células HT29 , Humanos , Necrosis/patología , Necrosis/prevención & control , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Células U937
16.
Nat Rev Cancer ; 24(5): 299-315, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38454135

RESUMEN

Most metastatic cancers remain incurable due to the emergence of apoptosis-resistant clones, fuelled by intratumour heterogeneity and tumour evolution. To improve treatment, therapies should not only kill cancer cells but also activate the immune system against the tumour to eliminate any residual cancer cells that survive treatment. While current cancer therapies rely heavily on apoptosis - a largely immunologically silent form of cell death - there is growing interest in harnessing immunogenic forms of cell death such as necroptosis. Unlike apoptosis, necroptosis generates second messengers that act on immune cells in the tumour microenvironment, alerting them of danger. This lytic form of cell death optimizes the provision of antigens and adjuvanticity for immune cells, potentially boosting anticancer treatment approaches by combining cellular suicide and immune response approaches. In this Review, we discuss the mechanisms of necroptosis and how it activates antigen-presenting cells, drives cross-priming of CD8+ T cells and induces antitumour immune responses. We also examine the opportunities and potential drawbacks of such strategies for exposing cancer cells to immunological attacks.


Asunto(s)
Muerte Celular Inmunogénica , Necroptosis , Neoplasias , Microambiente Tumoral , Humanos , Necroptosis/inmunología , Neoplasias/inmunología , Neoplasias/patología , Microambiente Tumoral/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Inmunoterapia/métodos
17.
Blood ; 118(3): 598-608, 2011 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-21613257

RESUMEN

The regulation of cellular survival and apoptosis is of critical importance for the immune system to maintain immune homeostasis and to establish tolerance. Here, we demonstrate that the immune specific cell surface molecule Toso exhibits antiapoptotic effects on death receptor signaling by a novel regulatory mechanism involving the adaptor kinase RIP1. The antiapoptotic function of Toso depends on RIP1 ubiquitination and involves the recruitment of the death adaptor FADD to a Toso/RIP1 protein complex. In response to CD95L and TNFα, Toso promotes the activation of MAPK and NF-κB signaling pathways. Because of this relative augmentation of survival versus apoptotic signals, Toso raises the threshold for death receptor-mediated apoptosis. Our analysis of Toso-deficient mice revealed that Toso is essential for TNFα-mediated liver damage. Furthermore, the antiapoptotic function of Toso could be blocked by a Toso-specific monoclonal antibody, opening up new therapeutic prospects for the treatment of immune disorders and hematologic malignancies.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/inmunología , Proteínas de la Membrana/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas de Unión al ARN/metabolismo , Transducción de Señal/inmunología , Ubiquitinación/fisiología , Animales , Anticuerpos Monoclonales/inmunología , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Supervivencia Celular/inmunología , Proteína Ligando Fas/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Tolerancia Inmunológica/inmunología , Células Jurkat , Hepatopatías/inmunología , Hepatopatías/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Unión al ARN/genética , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Receptor fas/metabolismo
18.
Cell Commun Signal ; 11: 76, 2013 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-24090154

RESUMEN

BACKGROUND: In apoptosis, proteolysis by caspases is the primary mechanism for both initiation and execution of programmed cell death (PCD). In contrast, the impact of proteolysis on the regulation and execution of caspase-independent forms of PCD (programmed necrosis, necroptosis) is only marginally understood. Likewise, the identity of the involved proteases has remained largely obscure. Here, we have investigated the impact of proteases in TNF-induced necroptosis. RESULTS: The serine protease inhibitor TPKC protected from TNF-induced necroptosis in multiple murine and human cells systems whereas inhibitors of metalloproteinases or calpain/cysteine and cathepsin proteases had no effect. A screen for proteins labeled by a fluorescent TPCK derivative in necroptotic cells identified HtrA2/Omi (a serine protease previously implicated in PCD) as a promising candidate. Demonstrating its functional impact, pharmacological inhibition or genetic deletion of HtrA2/Omi protected from TNF-induced necroptosis. Unlike in apoptosis, HtrA2/Omi did not cleave another protease, ubiquitin C-terminal hydrolase (UCH-L1) during TNF-induced necroptosis, but rather induced monoubiquitination indicative for UCH-L1 activation. Correspondingly, pharmacologic or RNA interference-mediated inhibition of UCH-L1 protected from TNF-induced necroptosis. We found that UCH-L1 is a mediator of caspase-independent, non-apoptotic cell death also in diseased kidney podocytes by measuring cleavage of the protein PARP-1, caspase activity, cell death and cell morphology. Indicating a role of TNF in this process, podocytes with stably downregulated UCH-L1 proved resistant to TNF-induced necroptosis. CONCLUSIONS: The proteases HtrA2/Omi and UCH-L1 represent two key components of TNF-induced necroptosis, validating the relevance of proteolysis not only for apoptosis, but also for caspase-independent PCD. Since UCH-L1 clearly contributes to the non-apoptotic death of podocytes, interference with the necroptotic properties of HtrA2/Omi and UCH-L1 may prove beneficial for the treatment of patients, e.g. in kidney failure.


Asunto(s)
Apoptosis/fisiología , Proteínas Mitocondriales/metabolismo , Serina Endopeptidasas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Células Cultivadas , Células HT29 , Serina Peptidasa A2 que Requiere Temperaturas Altas , Humanos , Células Jurkat , Ratones , Células 3T3 NIH , Podocitos/metabolismo , Ratas , Ratas Wistar
19.
Am J Respir Crit Care Med ; 185(2): 160-70, 2012 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-22246702

RESUMEN

RATIONALE: Platelet-activating factor (PAF) increases lung vascular permeability within minutes by activation of acid sphingomyelinase (ASM) and a subsequent nitric oxide (NO)-inhibitable and Ca(2+)-dependent loss in barrier function. OBJECTIVES: To elucidate the molecular mechanisms underlying this response. METHODS: In isolated perfused rat and mouse lungs, endothelial Ca(2+) concentration ([Ca(2+)](i)) was quantified by real-time fluorescence imaging, and caveolae of endothelial cells were isolated and probed for Ca(2+) entry channels. Regulation of transient receptor potential classical (TRPC) 6-mediated currents in lung endothelial cells was assessed by patch clamp technique. MEASUREMENTS AND MAIN RESULTS: PAF increased lung weight gain and endothelial [Ca(2+)](i). This response was abrogated by inhibitors of ASM or in ASM-deficient mice, and replicated by lung perfusion with exogenous ASM or C2-ceramide. PAF increased the caveolar abundance of TRPC6 channels, which was similarly blocked by ASM inhibition. PAF-induced increases in lung endothelial [Ca(2+)](i), vascular filtration coefficient, and edema formation were attenuated by the TRPC inhibitor SKF96365 and in TRPC6-deficient mice, whereas direct activation of TRPC6 replicated the [Ca(2+)](i) and edema response to PAF. The exogenous NO donor PapaNONOate or the cyclic guanosine 3',5'-monophosphate analog 8Br-cGMP blocked the endothelial [Ca(2+)](i) and permeability response to PAF, in that they directly blocked TRPC6 channels without interfering with their PAF-induced recruitment to caveolae. CONCLUSIONS: The present findings outline a new signaling cascade in the induction of PAF-induced lung edema, in that stimulation of ASM causes recruitment of TRPC6 channels to caveolae, thus allowing for Ca(2+) influx and subsequent increases in endothelial permeability that are amplified in the absence of endothelial NO synthesis.


Asunto(s)
Calcio/metabolismo , Permeabilidad Capilar , Endotelio Vascular/metabolismo , Pulmón/metabolismo , Factor de Activación Plaquetaria/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Endotelio Vascular/enzimología , Técnicas In Vitro , Pulmón/enzimología , Ratones , Óxido Nítrico/metabolismo , Ratas
20.
Proc Natl Acad Sci U S A ; 107(3): 1112-7, 2010 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-20080539

RESUMEN

The phospholipase neutral sphingomyelinase (N-SMase) has been recognized as a major mediator of processes such as inflammation, development and growth, differentiation and death of cells, as well as in diseases such as Alzheimer's, atherosclerosis, heart failure, ischemia/reperfusion damage, or combined pituitary hormone deficiency. Although activation of N-SMase by the proinflammatory cytokine TNF was described almost two decades ago, the underlying signaling pathway is unresolved. Here, we identify the Polycomb group protein EED (embryonic ectodermal development) as an interaction partner of nSMase2. In yeast, the N terminus of EED binds to the catalytic domain of nSMase2 as well as to RACK1, a protein that modulates the activation of nSMase2 by TNF in concert with the TNF receptor 1 (TNF-R1)-associated protein FAN. In mammalian cells, TNF causes endogenous EED to translocate from the nucleus and to colocalize and physically interact with both endogenous nSMase2 and RACK1. As a consequence, EED and nSMase2 are recruited to the TNF-R1.FAN.RACK1-complex in a timeframe concurrent with activation of nSMase2. After knockdown of EED by RNA interference, the TNF-dependent activation of nSMase2 is completely abrogated, identifying EED as a protein that both physically and functionally couples TNF-R1 to nSMase2, and which therefore represents the "missing link" that completes one of the last unresolved signaling pathways of TNF-R1.


Asunto(s)
Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas Represoras/metabolismo , Esfingomielina Fosfodiesterasa/metabolismo , Activación Enzimática , Células HeLa , Humanos , Complejo Represivo Polycomb 2
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA