Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 103
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Allergy ; 70(7): 764-74, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25833810

RESUMEN

BACKGROUND: Mastocytosis is characterized by the accumulation of mast cells (MCs) associated with activating mutations of KIT. Tumor necrosis factor-related apoptosis-inducing ligand receptors (TRAIL-Rs) are preferentially expressed on neoplastic cells and induce the extrinsic apoptotic pathway. Recent studies reported on the expression of TRAIL-Rs and TRAIL-induced apoptosis in cultured human MCs, which depend on stem cell factor (SCF)-induced or constitutive KIT activation. MATERIAL AND METHODS: We sought to further define the impact of TRAIL-Rs on MCs in vivo and in vitro. Using Cre/loxP recombination, we generated mice with MC-specific and ubiquitous knockout of TRAIL-R. In these mice, anaphylaxis and numbers of MCs were investigated. We also explored the expression and function of TRAIL-Rs in cultured murine and human MCs upon activation of KIT. By conducting immunofluorescence staining, we analyzed the expression of TRAIL-Rs in MCs infiltrating the bone marrow of patients with mastocytosis. RESULTS: MC-specific deletion of TRAIL-R was associated with a slight, but significant increase in anaphylaxis. Numbers of MCs in MC-specific knockouts of TRAIL-R were comparable to controls. Whereas cultured IL-3-dependent murine MCs from wild-type mice were resistant to TRAIL-induced apoptosis, SCF-stimulated MCs underwent apoptosis in response to TRAIL. Interestingly, activating KIT mutations also promoted sensitivity to TRAIL-mediated apoptosis in human MCs. In line with these findings, MCs infiltrating the bone marrow of patients with mastocytosis expressed TRAIL-R1. CONCLUSIONS: Activation of KIT regulates the function of TRAIL-Rs in MCs. TRAIL-R1 may represent an attractive diagnostic and therapeutic target in diseases associated with KIT mutations, such as mastocytosis.


Asunto(s)
Mastocitos/inmunología , Mastocitos/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Médula Ósea/inmunología , Médula Ósea/metabolismo , Médula Ósea/patología , Recuento de Células , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Mastocitosis/genética , Mastocitosis/inmunología , Mastocitosis/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación , Proteínas Proto-Oncogénicas c-kit/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Factor de Células Madre/metabolismo , Factor de Células Madre/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología
2.
Br J Cancer ; 111(10): 1955-64, 2014 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-25314064

RESUMEN

BACKGROUND: Current treatment strategies for head and neck cancer are associated with significant morbidity and up to 50% of patients relapse, highlighting the need for more specific and effective therapeutics. Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) and Smac mimetics (SMs) are promising anticancer agents, but their effect on head and neck squamous cell carcinoma (HNSCC) remains unknown. METHODS: We examined the response of a panel of nine HNSCC cell lines to TRAIL and SMs and investigated the mechanism of cell type-specific response by functional analysis. RESULTS: Head and neck cancer cell lines revealed a converse response pattern with three cell lines being highly sensitive to Smac-164 (SM) but resistant to TRAIL, whereas the other six were sensitive to TRAIL but resistant to SM. Distinct protein expression and activation patterns were found to be associated with susceptibility of HNSCC cell lines to TRAIL and SM. Tumour necrosis factor-related apoptosis-inducing ligand sensitivity was associated with high caspase-8 and Bid protein levels, and TRAIL-sensitive cell lines were killed via the type II extrinsic apoptotic pathway. Smac mimetic-sensitive cells expressed low levels of caspase-8 and Bid but had high TNF-α expression. Smac mimetic-induced cell death was associated with caspase-10 activation, suggesting that in the absence of caspase-8, caspase-10 mediates response to SM. Cotreatment with TNF-α sensitised the resistant cells to SM, demonstrating a decisive role for TNF-α-driven feedback loop in SM sensitivity. CONCLUSIONS: Tumour necrosis factor-related apoptosis-inducing ligand and SMs effectively kill HNSCC cell lines and therefore represent potential targeted therapeutics for head and neck cancer. Distinct molecular mechanisms determine the sensitivity to each agent, with levels of TNF-α, caspase-8, Bid and caspase-10 providing important predictive biomarkers of response to these agents.


Asunto(s)
Apoptosis/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Caspasa 10/metabolismo , Caspasa 8/metabolismo , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Triazoles/farmacología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Biomimética , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Proliferación Celular/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/metabolismo
3.
Cell Death Dis ; 15(5): 345, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38769311

RESUMEN

Treatment-naïve small cell lung cancer (SCLC) is typically susceptible to standard-of-care chemotherapy consisting of cisplatin and etoposide recently combined with PD-L1 inhibitors. Yet, in most cases, SCLC patients develop resistance to first-line therapy and alternative therapies are urgently required to overcome this resistance. In this study, we tested the efficacy of dinaciclib, an FDA-orphan drug and inhibitor of the cyclin-dependent kinase (CDK) 9, among other CDKs, in SCLC. Furthermore, we report on a newly developed, highly specific CDK9 inhibitor, VC-1, with tumour-killing activity in SCLC. CDK9 inhibition displayed high killing potential in a panel of mouse and human SCLC cell lines. Mechanistically, CDK9 inhibition led to a reduction in MCL-1 and cFLIP anti-apoptotic proteins and killed cells, almost exclusively, by intrinsic apoptosis. While CDK9 inhibition did not synergise with chemotherapy, it displayed high efficacy in chemotherapy-resistant cells. In vivo, CDK9 inhibition effectively reduced tumour growth and improved survival in both autochthonous and syngeneic SCLC models. Together, this study shows that CDK9 inhibition is a promising therapeutic agent against SCLC and could be applied to chemo-refractory or resistant SCLC.


Asunto(s)
Quinasa 9 Dependiente de la Ciclina , Indolizinas , Neoplasias Pulmonares , Compuestos de Piridinio , Carcinoma Pulmonar de Células Pequeñas , Quinasa 9 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 9 Dependiente de la Ciclina/metabolismo , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Carcinoma Pulmonar de Células Pequeñas/patología , Humanos , Animales , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Línea Celular Tumoral , Ratones , Compuestos de Piridinio/farmacología , Compuestos de Piridinio/uso terapéutico , Indolizinas/farmacología , Óxidos N-Cíclicos/farmacología , Apoptosis/efectos de los fármacos , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/uso terapéutico , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico
4.
Am J Transplant ; 13(11): 2797-804, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24103029

RESUMEN

Transplantation is invariably associated with ischemia-reperfusion injury (IRI), inflammation and rejection. Resultant cell death has morphological features of necrosis but programmed cell death has been synonymous with apoptosis until pathways of regulated necrosis (RN) have been described. The best-studied RN pathway, necroptosis, is triggered by perturbation of caspase-8-mediated apoptosis and depends on receptor-interacting protein kinases 1 and 3 (RIPK1/RIPK3) as well as mixed linage kinase domain like to form the necroptosome. The release of cytosolic content and cell death-associated molecular patterns (CDAMPs) can trigger innate and promote adaptive immune responses. Thus, the form of cell death can substantially influence alloimmunity and graft survival. Necroptosis is a key element of IRI, and RIPK1 interference by RN-specific inhibitors such as necrostatin-1 protects from IRI in kidney, heart and brain. Necroptosis may be a general mechanism in response to other forms of inflammatory organ injury, and will likely emerge as a promising target in solid organ transplantation. As second-generation RIPK1 and RIPK3 inhibitors become available, clinical trials for the prevention of delayed graft function and attenuation of allograft rejection-mediated injury will emerge. These efforts will accelerate upon further identification of critical necroptosis-triggering receptor(s).


Asunto(s)
Inmunidad Celular , Necrosis , Daño por Reperfusión/patología , Animales , Humanos
5.
Eur J Surg Oncol ; 49(11): 107096, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37801834

RESUMEN

BACKGROUND: The risk of an anastomotic leakage (AL) following Ivor-Lewis esophagectomy is increased in patients with calcifications of the aorta or a stenosis of the celiac trunc. Ischemic conditioning (ISCON) of the gastric conduit prior to esophagectomy is supposed to improve gastric vascularization at the anastomotic site. The prospective ISCON trial was conducted to proof the safety and feasibility of this strategy with partial gastric devascularization 14 days before esophagectomy in esophageal cancer patients with a compromised vascular status. This work reports the results from a translational project of the ISCON trial aimed to investigate variables of neo-angiogenesis. METHODS: Twenty esophageal cancer patients scheduled for esophagectomy were included in the ISCON trial. Serum samples (n = 11) were collected for measurement of biomarkers and biopsies (n = 12) of the gastric fundus were taken before and after ISCON of the gastric conduit. Serum samples were analyzed including 62 different cytokines. Vascularization of the gastric mucosa was assessed on paraffin-embedded sections stained against CD34 to detect the degree of microvascular density and vessel size. RESULTS: Between November 2019 and January 2022 patients were included in the ISCON Trial. While serum samples showed no differences regarding cytokine levels before and after ISCON biopsies of the gastric mucosa demonstrated a significant increase in microvascular density after ISCON as compared to the corresponding gastric sample before the intervention. CONCLUSION: The data prove that ISCON of the gastric conduit as esophageal substitute induces significant neo-angiogenesis in the gastric fundus which is considered as surrogate of an improved vascularization at the anastomotic site.


Asunto(s)
Neoplasias Esofágicas , Precondicionamiento Isquémico , Laparoscopía , Humanos , Esofagectomía/métodos , Estudios Prospectivos , Precondicionamiento Isquémico/métodos , Estómago/irrigación sanguínea , Isquemia , Neoplasias Esofágicas/cirugía , Neoplasias Esofágicas/patología
6.
Nat Med ; 5(2): 157-63, 1999 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9930862

RESUMEN

To evaluate the utility of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) as a cancer therapeutic, we created leucine zipper (LZ) forms of human (hu) and murine (mu) TRAIL to promote and stabilize the formation of trimers. Both were biologically active, inducing apoptosis of both human and murine target cells in vitro with similar specific activities. In contrast to the fulminant hepatotoxicity of LZ-huCD95L in vivo, administration of either LZ-huTRAIL or LZ-muTRAIL did not seem toxic to normal tissues of mice. Finally, repeated treatments with LZ-huTRAIL actively suppressed growth of the TRAIL-sensitive human mammary adenocarcinoma cell line MDA-231 in CB.17 (SCID) mice, and histologic examination of tumors from SCID mice treated with LZ-huTRAIL demonstrated clear areas of apoptotic necrosis within 9-12 hours of injection.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Glicoproteínas de Membrana/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Animales , Proteínas Reguladoras de la Apoptosis , Relación Dosis-Respuesta a Droga , Proteína Ligando Fas , Humanos , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/síntesis química , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones SCID , Conformación Proteica , Ligando Inductor de Apoptosis Relacionado con TNF , Células Tumorales Cultivadas , Factor de Necrosis Tumoral alfa/administración & dosificación , Factor de Necrosis Tumoral alfa/síntesis química
7.
J Exp Med ; 190(8): 1103-14, 1999 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-10523608

RESUMEN

Herpes simplex virus type 1 (HSV1), a large DNA-containing virus, is endemic in all human populations investigated. After infection of mucocutaneous surfaces, HSV1 establishes a latent infection in nerve cells. Recently, it was demonstrated that HSV1 can also infect activated T lymphocytes. However, the consequences of T cell infection for viral pathogenesis and immunity are unknown. We have observed that in contrast to the situation in human fibroblasts, in human T cell lines antigen presentation by major histocompatibility complex class I molecules is not blocked after HSV1 infection. Moreover, HSV1 infection of T cells results in rapid elimination of antiviral T cells by fratricide. To dissect the underlying molecular events, we used a transgenic mouse model of HSV1 infection to demonstrate that CD95 (Apo-1, Fas)-triggered apoptosis is essential for HSV1-induced fratricide, whereas tumor necrosis factor (TNF) also contributes to this phenomenon but to a lesser extent. By contrast, neither TRAIL (TNF-related apoptosis-inducing ligand) nor perforin were involved. Finally, we defined two mechanisms associated with HSV1-associated fratricide of antiviral T cells: (a) T cell receptor-mediated upregulation of CD95 ligand and (b) a viral "competence-to-die" signal that renders activated T lymphocytes susceptible to CD95 signaling. We propose that induction of fratricide is an important immune evasion mechanism of HSV1, helping the virus to persist in the host organism throughout its lifetime.


Asunto(s)
Herpesvirus Humano 1/inmunología , Activación de Linfocitos/inmunología , Linfocitos T Citotóxicos/virología , Animales , Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis , Proteína Ligando Fas , Citometría de Flujo , Genes MHC Clase I/inmunología , Herpesvirus Humano 1/patogenicidad , Humanos , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Transgénicos , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF , Factor de Necrosis Tumoral alfa/inmunología , Receptor fas/inmunología
8.
J Exp Med ; 182(5): 1223-30, 1995 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-7595193

RESUMEN

Apoptosis occurs in the normal liver and in various forms of liver disease. The CD95 (APO-1/Fas) (CD95) receptor mediates apoptosis, and liver cells in animal models are acutely sensitive to apoptosis initiated by this receptor. We have used primary human hepatocytes as a model system to investigate CD95-mediated apoptotic liver damage. Treatment of fresh human hepatocytes with low concentrations of agonistic antibodies against CD95 resulted in apoptosis of > 95% of the cultured liver cells within 4 and 7.5 h. Immunohistology of a panel of explanted liver tissues revealed that hepatocytes in normal livers (n = 5) and in alcoholic cirrhosis (n = 13) expressed low constitutive levels of CD95. CD95 receptor expression was highly elevated in hepatocytes in hepatitis B virus-related cirrhosis (n = 9) and in acute liver failure (n = 8). By in situ hybridization CD95 ligand messenger RNA expression was absent in normal liver but detected at high levels in livers with ongoing liver damage. In cases of hepatitis B virus-related cirrhosis and acute hepatic failure, ligand expression was found primarily in areas with lymphocytic infiltration. In contrast, in patients with alcoholic liver damage, high CD95 ligand messenger RNA expression was found in hepatocytes. These findings suggest that liver destruction in hepatitis B may primarily involve killing of hepatocytes by T lymphocytes using the CD95 receptor-ligand system. In alcoholic liver damage, death of hepatocytes might occur by fratricide and paracrine or autocrine mechanisms mediated by the hepatocytes themselves.


Asunto(s)
Apoptosis/fisiología , Hígado/patología , Glicoproteínas de Membrana/fisiología , Linfocitos T Citotóxicos/inmunología , Receptor fas/fisiología , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Células Cultivadas , Proteína Ligando Fas , Hepatitis Viral Humana/complicaciones , Hepatitis Viral Humana/patología , Humanos , Hibridación Fluorescente in Situ , Cirrosis Hepática/etiología , Cirrosis Hepática/patología , Cirrosis Hepática Alcohólica/patología , Fallo Hepático/patología , Activación de Linfocitos , Glicoproteínas de Membrana/biosíntesis , Glicoproteínas de Membrana/genética , ARN Mensajero/análisis , Receptor fas/inmunología
9.
J Exp Med ; 186(7): 1165-70, 1997 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-9314565

RESUMEN

TRAIL-R3, a new member of the TRAIL receptor family, has been cloned and characterized. TRAIL-R3 encodes a 299 amino acid protein with 58 and 54% overall identity to TRAIL-R1 and -R2, respectively. Transient expression and quantitative binding studies show TRAIL-R3 to be a plasma membrane-bound protein capable of high affinity interaction with the TRAIL ligand. The TRAIL-R3 gene maps to human chromosome 8p22-21, clustered with the genes encoding two other TRAIL receptors. In contrast to TRAIL-R1 and -R2, this receptor shows restricted expression, with transcripts detectable only in peripheral blood lymphocytes and spleen. The structure of TRAIL-R3 is unique when compared to the other TRAIL receptors in that it lacks a cytoplasmic domain and appears to be glycosyl-phosphatidylinositol-linked. Moreover, unlike TRAIL-R1 and -R2, in a transient overexpression system TRAIL-R3 does not induce apoptosis.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Receptores de Superficie Celular/química , Receptores de Superficie Celular/metabolismo , Receptores del Factor de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/metabolismo , Secuencia de Aminoácidos , Apoptosis/fisiología , Proteínas Reguladoras de la Apoptosis , Secuencia de Bases , Northern Blotting , Mapeo Cromosómico , Cromosomas Humanos Par 8/genética , Clonación Molecular , Proteínas Ligadas a GPI , Regulación de la Expresión Génica , Glicosilfosfatidilinositoles , Humanos , Fragmentos Fc de Inmunoglobulinas/genética , Células Jurkat , Linfocitos/química , Linfocitos/metabolismo , Glicoproteínas de Membrana/farmacología , Datos de Secuencia Molecular , Unión Proteica , Receptores de Superficie Celular/genética , Miembro 10c de Receptores del Factor de Necrosis Tumoral , Proteínas Recombinantes de Fusión/farmacología , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Ligando Inductor de Apoptosis Relacionado con TNF , Receptores Señuelo del Factor de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/farmacología
10.
Br J Cancer ; 102(12): 1707-16, 2010 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-20461078

RESUMEN

BACKGROUND: XIAP (X-linked inhibitor of apoptosis protein) is an anti-apoptotic protein exerting its activity by binding and suppressing caspases. As XIAP is overexpressed in several tumours, in which it apparently contributes to chemoresistance, and because its activity in vivo is antagonised by second mitochondria-derived activator of caspase (SMAC)/direct inhibitor of apoptosis-binding protein with low pI, small molecules mimicking SMAC (so called SMAC-mimetics) can potentially overcome tumour resistance by promoting apoptosis. METHODS: Three homodimeric compounds were synthesised tethering a monomeric SMAC-mimetic with different linkers and their affinity binding for the baculoviral inhibitor repeats domains of XIAP measured by fluorescent polarisation assay. The apoptotic activity of these molecules, alone or in combination with tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) and/or Bortezomib, was tested in melanoma cell lines by MTT viability assays and western blot analysis of activated caspases. RESULTS: We show that in melanoma cell lines, which are typically resistant to chemotherapeutic agents, XIAP knock-down sensitises cells to TRAIL treatment in vitro, also favouring the accumulation of cleaved caspase-8. We also describe a new series of 4-substituted azabicyclo[5.3.0]alkane monomeric and dimeric SMAC-mimetics that target various members of the IAP family and powerfully synergise at submicromolar concentrations with TRAIL in inducing cell death. Finally, we show that the simultaneous administration of newly developed SMAC-mimetics with Bortezomib potently triggers apoptosis in a melanoma cell line resistant to the combined effect of SMAC-mimetics and TRAIL. CONCLUSION: Hence, the newly developed SMAC-mimetics effectively synergise with TRAIL and Bortezomib in inducing cell death. These findings warrant further preclinical studies in vivo to verify the anticancer effectiveness of the combination of these agents.


Asunto(s)
Ácidos Borónicos/farmacología , Muerte Celular/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/farmacología , Melanoma/tratamiento farmacológico , Proteínas Mitocondriales/farmacología , Pirazinas/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Proteínas Reguladoras de la Apoptosis , Bortezomib , Caspasa 8/metabolismo , Línea Celular Tumoral , Regulación hacia Abajo , Interacciones Farmacológicas , Sinergismo Farmacológico , Técnicas de Silenciamiento del Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular/administración & dosificación , Proteínas Mitocondriales/administración & dosificación , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación
11.
Cell Death Differ ; 15(11): 1681-90, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18583989

RESUMEN

Although nuclear factor-kappaB (NF-kappaB) usually exerts anti-apoptotic activity, upon activation by interleukin-1 (IL-1) it enhances ultraviolet-B radiation (UVB)-induced apoptosis. This paradoxical effect is associated with NF-kappaB-dependent pronounced secretion of tumour necrosis factor-alpha (TNF) which activates TNF-R1 in an autocrine fashion to enhance UVB-induced apoptosis. We demonstrate that sustained TNF transcription in UVB+IL-1-treated cells involves complete abrogation of the negative feedback loop of NF-kappaB preventing IkappaBalpha resynthesis, hence allowing uncontrolled NF-kappaB activity. We show that IkappaBalpha is not transcriptionally inhibited but resynthesized protein is immediately marked for degradation due to persistent inhibitor of kappaB kinasebeta (IKKbeta) activity. Continuous IKKbeta phosphorylation and activation is caused by UVB-mediated inhibition of the phosphatase PP2A. This study demonstrates that the cellular response to different NF-kappaB activators may be converted to the opposite reaction when both stimuli act in concert. Our data shed new light on the significance of negative feedback regulation of NF-kappaB and identifies PP2A as the key regulator of this process.


Asunto(s)
Apoptosis/efectos de la radiación , Retroalimentación Fisiológica/efectos de la radiación , FN-kappa B/metabolismo , Proteína Fosfatasa 2/metabolismo , Rayos Ultravioleta , Apoptosis/efectos de los fármacos , Comunicación Autocrina/efectos de los fármacos , Comunicación Autocrina/efectos de la radiación , Línea Celular Tumoral , Retroalimentación Fisiológica/efectos de los fármacos , Humanos , Quinasa I-kappa B/metabolismo , Proteínas I-kappa B/antagonistas & inhibidores , Interleucina-1/farmacología , Modelos Biológicos , Inhibidor NF-kappaB alfa , Fosforilación/efectos de los fármacos , Fosforilación/efectos de la radiación , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de la radiación , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/efectos de la radiación , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Transcripción Genética/efectos de los fármacos , Transcripción Genética/efectos de la radiación , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
12.
Cell Death Differ ; 15(4): 762-72, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18202702

RESUMEN

We investigated the mechanisms involved in the resistance to cell death observed in epithelial cancers. Here, we identify that primary epithelial cancer cells from colon, breast and lung carcinomas express high levels of the antiapoptotic proteins PED, cFLIP, Bcl-xL and Bcl-2. These cancer cells produced interleukin-4 (IL-4), which amplified the expression levels of these antiapoptotic proteins and prevented cell death induced upon exposure to TRAIL or other drug agents. IL-4 blockade resulted in a significant decrease in the growth rate of epithelial cancer cells and sensitized them, both in vitro and in vivo, to apoptosis induction by TRAIL and chemotherapy via downregulation of the antiapoptotic factors PED, cFLIP, Bcl-xL and Bcl-2. Furthermore, we provide evidence that exogenous IL-4 was able to upregulate the expression levels of these antiapoptotic proteins and potently stabilized the growth of normal epithelial cells rendering them apoptosis resistant. In conclusion, IL-4 acts as an autocrine survival factor in epithelial cells. Our results indicate that inhibition of IL-4/IL-4R signaling may serve as a novel treatment for epithelial cancers.


Asunto(s)
Apoptosis , Comunicación Autocrina , Neoplasias de la Mama/metabolismo , Carcinoma/metabolismo , Neoplasias del Colon/metabolismo , Resistencia a Antineoplásicos , Interleucina-4/metabolismo , Neoplasias Pulmonares/metabolismo , Adulto , Anciano , Anticuerpos Monoclonales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Carcinoma/tratamiento farmacológico , Carcinoma/patología , Muerte Celular , Proliferación Celular , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Interleucina-4/inmunología , Subunidad alfa del Receptor de Interleucina-4/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Factores de Tiempo , Células Tumorales Cultivadas , Regulación hacia Arriba , Proteína bcl-X/metabolismo
13.
J Cell Biol ; 127(1): 15-20, 1994 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-7523418

RESUMEN

Apoptosis is the predominant form of cell death and occurs under a variety of physiological and pathological conditions. Cells undergoing apoptotic cell death reveal a characteristic sequence of cytological alterations including membrane blebbing and nuclear and cytoplasmic condensation. Activation of an endonuclease which cleaves genomic DNA into internucleosomal DNA fragments is considered to be the hallmark of apoptosis. However, no clear evidence exists that DNA degradation plays a primary and causative role in apoptotic cell death. Here we show that cells enucleated with cytochalasin B still undergo apoptosis induced either by treatment with menadione, an oxidant quinone compound, or by triggering APO-1/Fas, a cell surface molecule involved in physiological cell death. Incubation of enucleated cells with the agonistic monoclonal anti-APO-1 antibody revealed the key morphological features of apoptosis. Moreover, in non-enucleated cells inhibitors of endonuclease blocked DNA fragmentation, but not cell death induced by anti-APO-1. These data suggest that DNA degradation and nuclear signaling are not required for induction of apoptotic cell death.


Asunto(s)
Apoptosis/fisiología , Núcleo Celular/fisiología , ADN/metabolismo , Animales , Anticuerpos Monoclonales , Antígenos de Superficie/fisiología , Apoptosis/efectos de los fármacos , Ácido Aurintricarboxílico/farmacología , Membrana Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Supervivencia Celular , Desoxirribonucleasa I/antagonistas & inhibidores , Células L , Ratones , Vitamina K/farmacología , Receptor fas
14.
Cell Death Differ ; 26(12): 2808-2809, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31395960

RESUMEN

Authors have only now noticed that in the Figure 3a, the immunohistochemical analysis of IL-4Rα on paraffin-embedded sections from breast is incorrect: IL-4 from breast was duplicated and used for the IL-4Rα staining. The correct Figure 3a has been included in the amendment to this paper.An amendment to this paper has been published and can be accessed via a link at the top of the paper.

15.
Cell Death Differ ; 13(10): 1740-51, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16470224

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selectively induces apoptosis in many transformed cells, suggesting TRAIL as an ideal candidate for cancer gene therapy. A main obstacle in cancer therapy is intrinsic or acquired therapy resistance of malignant cells. To study induction of resistance against TRAIL, we generated lentiviral vectors allowing efficient TRAIL expression and apoptosis induction in a variety of human cancer cell lines. Within days upon TRAIL overexpression, cells became resistant towards TRAIL, but not to CD95 ligation or DNA damage by cisplatin. Cell surface expression of TRAIL receptors 1 and 2 was completely abrogated in resistant cells due to intracellular retention of the receptors by TRAIL. SiRNA directed against TRAIL resensitized the resistant cells by restoring cell surface expression of TRAIL receptors. These findings represent a novel resistance mechanism towards TRAIL, specifically caused by TRAIL overexpression, and question the use of TRAIL expression in tumor-cell targeting gene therapy.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Lentivirus/genética , Glicoproteínas de Membrana/genética , Receptores del Factor de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/genética , Apoptosis , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Secuencia de Bases , Línea Celular Tumoral , Cisplatino/farmacología , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte , Resistencia a Antineoplásicos , Retículo Endoplásmico/metabolismo , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Aparato de Golgi/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Células Jurkat , Glicoproteínas de Membrana/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , ARN Interferente Pequeño/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Receptores del Factor de Necrosis Tumoral/genética , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF , Transducción Genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
16.
J Clin Invest ; 99(3): 403-13, 1997 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-9022073

RESUMEN

Chemotherapeutic drugs are cytotoxic by induction of apoptosis in drug-sensitive cells. We investigated the mechanism of bleomycin-induced cytotoxicity in hepatoma cells. At concentrations present in the sera of patients during therapy, bleomycin induced transient accumulation of nuclear wild-type (wt) p53 and upregulated expression of cell surface CD95 (APO-1/Fas) receptor in hepatoma cells carrying wt p53 (HepG2). Bleomycin did not increase CD95 in hepatoma cells with mutated p53 (Huh7) or in hepatoma cells which were p53-/- (Hep3B). In addition, sensitivity towards CD95-mediated apoptosis was also increased in wt p53 positive HepG2 cells. Microinjection of wt p53 cDNA into HepG2 cells had the same effect. In contrast, bleomycin did not enhance susceptibility towards CD95-mediated apoptosis in Huh7 and in Hep3B cells. Furthermore, bleomycin treatment of HepG2 cells increased CD95 ligand (CD95L) mRNA expression. Most notably, bleomycin-induced apoptosis in HepG2 cells was almost completely inhibited by antibodies which interfere with CD95 receptor/ligand interaction. These data suggest that apoptosis induced by bleomycin is mediated, at least in part, by p53-dependent stimulation of the CD95 receptor/ligand system. The same applies to other anti-cancer drugs such as cisplatin and methotrexate. These data may have major consequences for drug treatment of cancer and the explanation of drug sensitivity and resistance.


Asunto(s)
Antimetabolitos Antineoplásicos/farmacología , Antimetabolitos Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Bleomicina/farmacología , Bleomicina/toxicidad , Carcinoma Hepatocelular/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica , Genes p53 , Glicoproteínas de Membrana/inmunología , Receptor fas/inmunología , Anticuerpos Bloqueadores/inmunología , Antimetabolitos Antineoplásicos/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Antineoplásicos/toxicidad , Apoptosis/genética , Apoptosis/inmunología , Bleomicina/uso terapéutico , Western Blotting , Células Cultivadas , Cisplatino/farmacología , Cisplatino/uso terapéutico , Cisplatino/toxicidad , Pruebas Inmunológicas de Citotoxicidad , Fragmentación del ADN , ADN Complementario/genética , Resistencia a Antineoplásicos , Proteína Ligando Fas , Humanos , Inmunohistoquímica , Hibridación in Situ , Glicoproteínas de Membrana/genética , Metotrexato/farmacología , Metotrexato/uso terapéutico , Metotrexato/toxicidad , Microinyecciones , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , ARN Mensajero/biosíntesis
17.
Cancer Gene Ther ; 14(3): 316-26, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17186015

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising anticancer agent, which selectively induces apoptosis in many transformed cells without apparent toxic side effects in normal tissue. We recently described the construction and characterization of a lentiviral vector for expression of TRAIL. In this report, we evaluate its suitability for therapeutic application. In vitro, we observed specific induction of apoptosis upon transduction in human lung cancer cells. Cell death was partially dependent on successful integration and TRAIL expression by the vectors, but was to some extent mediated by protein carryover, as we found TRAIL protein associated with virus particles. Transduction of subcutaneously growing lung tumors on nude mice with lentiviral TRAIL mediated a transient suppression of tumor growth. Analysis of tumor sections revealed that transduction efficiency of lentiviral control vector but not of lentiviral TRAIL vector was high. This was because of the direct cytotoxic activity of recombinant TRAIL present in viral particles, which prevented efficient tumor transduction. These data therefore suggest that enveloped viral vectors constitutively expressing TRAIL are well suited for ex vivo applications, such as the transduction of tumor-homing cells, but may have a lower effect when used directly for the transduction of tumor cells in vivo.


Asunto(s)
Apoptosis , Carcinoma de Pulmón de Células no Pequeñas/terapia , Terapia Genética , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Células Cultivadas , Femenino , Humanos , Riñón/metabolismo , Ligandos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Transducción Genética , Factor de Necrosis Tumoral alfa/farmacología , Virión/metabolismo
18.
Cancer Res ; 60(11): 3051-7, 2000 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-10850456

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a member of the tumor necrosis factor family and has recently been shown to exert tumoricidal activity in vivo in the absence of any observable toxicity. The signaling pathways triggered by TRAIL stimulation and the mechanisms involved in resistance against TRAIL-mediated apoptosis are still poorly defined. We show here that TRAIL-induced apoptosis involves late dissipation of mitochondrial membrane potential (delta psi(m)) and cytochrome c release. These events follow activation of caspase-8 and caspase-3 and induction of DNA fragmentation. In addition, caspase-8-deficient cells are resistant against TRAIL-induced apoptosis, and inhibition of caspase-8 but not caspase-9 prevents mitochondrial permeability transition and apoptosis. In contrast, various Bcl-2- or Bcl-xL-overexpressing tumor cell lines are sensitive to TRAIL-induced apoptosis; however, they show a delay in TRAIL-induced mitochondrial permeability transition compared with control transfectants. This indicates that TRAIL-induced apoptosis depends on caspase-8 activation rather than on the disruption of mitochondrial integrity. Because most chemotherapeutic drugs used in the treatment of malignancies lead to apoptosis primarily by engagement of the mitochondrial proapoptotic machinery, we tested whether drug-resistant tumor cells retain sensitivity for TRAIL-induced apoptosis. Tumor cells overexpressing Bcl-2 or Bcl-xL become resistant to apoptosis induced by the chemotherapeutic drug etoposide. However, these cells are not protected or are only marginally protected against TRAIL-induced apoptosis. Thus, TRAIL may still kill tumors that have acquired resistance to chemotherapeutic drugs by overexpression of Bcl-2 or Bcl-xL. These data will influence future treatment strategies involving TRAIL.


Asunto(s)
Resistencia a Antineoplásicos , Glicoproteínas de Membrana/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Antineoplásicos Fitogénicos/metabolismo , Apoptosis , Proteínas Reguladoras de la Apoptosis , Caspasa 3 , Caspasa 8 , Caspasa 9 , Caspasas/metabolismo , Supervivencia Celular/efectos de los fármacos , Grupo Citocromo c/metabolismo , Fragmentación del ADN , Relación Dosis-Respuesta a Droga , Activación Enzimática , Etopósido/metabolismo , Citometría de Flujo , Humanos , Células Jurkat , Mitocondrias/metabolismo , Fosfatidilserinas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF , Factores de Tiempo , Células Tumorales Cultivadas , Proteína bcl-X
19.
Cancer Res ; 60(3): 553-9, 2000 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-10676636

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been shown to exert potent cytotoxic activity against many tumor cell lines but not against normal cells. It has been hypothesized that this difference in TRAIL sensitivity between normal and transformed cells might be due to the expression of the non-death-inducing TRAIL receptors (TRAIL-R) TRAIL-R3 and TRAIL-R4, presumably by competition for limited amounts of TRAIL. To assess the regulation of resistance versus sensitivity to TRAIL in primary as well as transformed keratinocytes, we examined TRAIL sensitivity, TRAIL receptor expression, and intracellular signaling events induced by TRAIL. Although TRAIL induced apoptosis in primary as well as transformed keratinocytes, a marked difference in sensitivity could be observed with primary keratinocytes (PK) being 5-fold less sensitive to TRAIL than transformed keratinocytes (TK). Yet both cell types exhibited similar TRAIL receptor surface expression, suggesting that expression of TRAIL-R3 and TRAIL-R4 may not be the main regulator of sensitivity to TRAIL. Biochemical analysis of the signaling events induced by TRAIL revealed that PK could be sensitized for TRAIL and, similarly, for TRAIL-R1- and TRAIL-R2-specific apoptosis by pretreatment of the cells with cycloheximide (CHX). This sensitization concomitantly resulted in processing of caspase-8, which did not occur in TRAIL-resistant PK. These data indicate that an early block of TRAIL-induced apoptosis was present in PK compared with TK or PK treated with CHX. Interestingly, cellular FLICE inhibitory protein (cFLIP) levels, high in PK and low in TK and several other squamous cell carcinoma cell lines, decreased rapidly after treatment of PK with CHX, correlating with the increase in TRAIL sensitivity and caspase-8 processing. Furthermore, ectopic expression of cFLIP long (cFLIP(L)) in TK by transfection with a cFLIP(L) expression vector resulted in resistance to TRAIL-mediated apoptosis of these cells. Thus, our results demonstrate that TRAIL sensitivity in PK is primarily regulated at the intracellular level rather than at the receptor level.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular , Queratinocitos/efectos de los fármacos , Glicoproteínas de Membrana/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD , Proteínas Portadoras/análisis , Caspasa 8 , Caspasa 9 , Inhibidores de Caspasas , Células Cultivadas , Cicloheximida/farmacología , Proteínas Ligadas a GPI , Humanos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF , Receptores del Factor de Necrosis Tumoral/análisis , Miembro 10c de Receptores del Factor de Necrosis Tumoral , Ligando Inductor de Apoptosis Relacionado con TNF , Receptores Señuelo del Factor de Necrosis Tumoral
20.
Cell Death Differ ; 23(5): 733-47, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26943322

RESUMEN

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), also known as Apo-2 ligand (Apo2L), is a member of the TNF cytokine superfamily. By cross-linking TRAIL-Receptor (TRAIL-R) 1 or TRAIL-R2, also known as death receptors 4 and 5 (DR4 and DR5), TRAIL has the capability to induce apoptosis in a wide variety of tumor cells while sparing vital normal cells. The discovery of this unique property among TNF superfamily members laid the foundation for testing the clinical potential of TRAIL-R-targeting therapies in the cancer clinic. To date, two of these therapeutic strategies have been tested clinically: (i) recombinant human TRAIL and (ii) antibodies directed against TRAIL-R1 or TRAIL-R2. Unfortunately, however, these TRAIL-R agonists have basically failed as most human tumors are resistant to apoptosis induction by them. It recently emerged that this is largely due to the poor agonistic activity of these agents. Consequently, novel TRAIL-R-targeting agents with increased bioactivity are currently being developed with the aim of rendering TRAIL-based therapies more active. This review summarizes these second-generation novel formulations of TRAIL and other TRAIL-R agonists, which exhibit enhanced cytotoxic capacity toward cancer cells, thereby providing the potential of being more effective when applied clinically than first-generation TRAIL-R agonists.


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Ligando Inductor de Apoptosis Relacionado con TNF/agonistas , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Apoptosis/efectos de los fármacos , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Relación Estructura-Actividad , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA