Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Am J Transplant ; 17(4): 1125-1128, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27931087

RESUMEN

We report the case of a 40-year-old woman who recovered from a diffuse metastatic renal cell carcinoma that developed from a kidney allograft. She was successfully treated by the induction of tumor rejection. Immunosuppression was discontinued, and transplant nephrectomy was deliberately delayed based on the expectation that the tumor mass would trigger the alloimmune response, which was stimulated with pegylated interferon-α-2a. Three years later, the patient remained in complete remission. Despite this severe context, the present case shows that the poor prognosis of allograft metastatic renal cell carcinoma could be dramatically reversed by taking advantage of the donor tumor origin to actively induce a specific alloimmune rejection of the tumor.


Asunto(s)
Carcinoma de Células Renales/tratamiento farmacológico , Interferón-alfa/uso terapéutico , Neoplasias Renales/tratamiento farmacológico , Trasplante de Riñón/efectos adversos , Polietilenglicoles/uso terapéutico , Adulto , Antivirales/uso terapéutico , Carcinoma de Células Renales/etiología , Carcinoma de Células Renales/secundario , Femenino , Humanos , Neoplasias Renales/etiología , Neoplasias Renales/patología , Proteínas Recombinantes/uso terapéutico , Inducción de Remisión , Trasplante Homólogo
2.
Rev Med Interne ; 37(5): 307-20, 2016 May.
Artículo en Francés | MEDLINE | ID: mdl-26899776

RESUMEN

PURPOSE: To develop French recommendations about the management of vaccinations, the screening of cervical cancer and the prevention of pneumocystis pneumonia in systemic lupus erythematosus (SLE). METHODS: Thirty-seven experts qualified in internal medicine, rheumatology, dermatology, nephrology and pediatrics have selected recommendations from a list of proposition based on available data from the literature. For each recommendation, the level of evidence and the level of agreement among the experts were specified. RESULTS: Inactivated vaccines do not cause significant harm in SLE patients. Experts recommend that lupus patient should receive vaccinations accordingly to the recommendations and the schedules for the general public. Pneumococcal vaccination is recommended for all SLE patients. Influenza vaccination is recommended for immunosuppressed SLE patients. Live attenuated vaccines should be avoided in immunosuppressed patients. Yet, recent works suggest that they can be considered in mildly immunosuppressed patients. Experts have recommended a cervical cytology every year for immunosuppressed patients. No consensus was obtained for the prevention of pneumocystis pneumonia. CONCLUSION: These recommendations can be expected to improve clinical practice uniformity and, in the longer term, to optimize the management of SLE patients.


Asunto(s)
Testimonio de Experto , Control de Infecciones/normas , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/terapia , Guías de Práctica Clínica como Asunto , Adolescente , Adulto , Francia , Humanos , Huésped Inmunocomprometido , Control de Infecciones/métodos , Infecciones/diagnóstico , Lupus Eritematoso Sistémico/diagnóstico , Lupus Eritematoso Sistémico/inmunología , Literatura de Revisión como Asunto , Vacunación/normas , Adulto Joven
3.
Rev Med Interne ; 36(6): 372-80, 2015 Jun.
Artículo en Francés | MEDLINE | ID: mdl-25455954

RESUMEN

PURPOSE: To develop French recommendations about screening and management of cardiovascular risk factors in systemic lupus erythematosus (SLE). METHODS: Thirty-nine experts qualified in internal medicine, rheumatology and nephrology have selected recommendations from a list developed based on evidence from the literature. For each recommendation, the level of evidence and the level of agreement among the experts were specified. RESULTS: Experts recommended an annual screening of cardiovascular risk factors in SLE. Statins should be prescribed for primary prevention in SLE patients based on the level of LDL-cholesterol and the number of cardiovascular risk factors, considering SLE as an additional risk factor. For secondary prevention, experts have agreed on an LDL-cholesterol target of <0.7 g/L. Hypertension should be managed according to the 2013 European guidelines, using renin-angiotensin system blockers as first line agents in case of renal involvement. Aspirin can be prescribed in patients with high cardiovascular risk or with antiphospholipid antibodies. CONCLUSION: These recommendations about the screening and management of cardiovascular risk factors in SLE can be expected to improve clinical practice uniformity and, in the longer term, to optimize the management of SLE patients.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Lupus Eritematoso Sistémico/complicaciones , Tamizaje Masivo/métodos , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/tratamiento farmacológico , Medicina Basada en la Evidencia , Testimonio de Experto , Guías como Asunto , Humanos , Factores de Riesgo , Prevención Secundaria
5.
FASEB J ; 15(3): 758-67, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11259394

RESUMEN

The complete AIF cDNA comprising the amino-terminal mitochondrial localization sequence (MLS) and the oxidoreductase domain has been fused in its carboxyl terminus to enhanced green fluorescent protein (GFP), thereby engineering an AIF-GFP fusion protein that is selectively targeted to the mitochondrial intermembrane space. Upon induction of apoptosis, the AIF-GFP protein translocates together with cytochrome c (Cyt-c) to the extramitochondrial compartment. Microinjection of recombinant AIF leads to the release of AIF-GFP and Cyt-c-GFP, indicating that ectopic AIF can favor permeabilization of the outer mitochondrial membrane. These mitochondrial effects of AIF are caspase independent, whereas the Cyt-c-microinjection induced translocation of AIF-GFP and Cyt-c-GFP is suppressed by the pan-caspase inhibitor Z-VAD.fmk. Upon prolonged culture, transfection-enforced overexpression of AIF results in spontaneous translocation of AIF-GFP from mitochondria, nuclear chromatin condensation, and cell death. These effects are caspase independent and do not rely on the oxidoreductase function of AIF. Spontaneous AIF-GFP translocation and subsequent nuclear apoptosis can be retarded by overexpression of a Bcl-2 protein selectively targeted to mitochondria, but not by a Bcl-2 protein targeted to the endoplasmic reticulum. Overexpression of a mutant AIF protein in which the MLS has been deleted (AIF Delta 1-100) results in the primary cytosolic accumulation of AIF. AIF Delta 1-100-induced cell death is suppressed by neither Z-VAD.fmk or by Bcl-2. Thus, extramitochondrially targeted AIF is a dominant cell death inducer.


Asunto(s)
Apoptosis/fisiología , Flavoproteínas/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Señales de Clasificación de Proteína/genética , Transporte de Proteínas/fisiología , Transporte Activo de Núcleo Celular , Secuencia de Aminoácidos , Animales , Factor Inductor de la Apoptosis , Células COS , Clonación Molecular , Cricetinae , Grupo Citocromo c/genética , Grupo Citocromo c/metabolismo , Flavoproteínas/genética , Colorantes Fluorescentes/metabolismo , Genes Reporteros/genética , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/metabolismo , Proteínas de la Membrana/genética , Microinyecciones , Microscopía Confocal , Datos de Secuencia Molecular , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Ratas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Alineación de Secuencia , Factores de Tiempo , Transfección
6.
J Exp Med ; 192(4): 571-80, 2000 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-10952727

RESUMEN

Apaf-1(-/-) or caspase-3(-/-) cells treated with a variety of apoptosis inducers manifest apoptosis-associated alterations including the translocation of apoptosis-inducing factor (AIF) from mitochondria to nuclei, large scale DNA fragmentation, and initial chromatin condensation (stage I). However, when compared with normal control cells, Apaf-1(-/-) or caspase-3(-/-) cells fail to exhibit oligonucleosomal chromatin digestion and a more advanced pattern of chromatin condensation (stage II). Microinjection of such cells with recombinant AIF only causes peripheral chromatin condensation (stage I), whereas microinjection with activated caspase-3 or its downstream target caspase-activated DNAse (CAD) causes a more pronounced type of chromatin condensation (stage II). Similarly, when added to purified HeLa nuclei, AIF causes stage I chromatin condensation and large-scale DNA fragmentation, whereas CAD induces stage II chromatin condensation and oligonucleosomal DNA degradation. Furthermore, in a cell-free system, concomitant neutralization of AIF and CAD is required to suppress the nuclear DNA loss caused by cytoplasmic extracts from apoptotic wild-type cells. In contrast, AIF depletion alone suffices to suppress the nuclear DNA loss contained in extracts from apoptotic Apaf-1(-/-) or caspase-3(-/-) cells. As a result, at least two redundant parallel pathways may lead to chromatin processing during apoptosis. One of these pathways involves Apaf-1 and caspases, as well as CAD, and leads to oligonucleosomal DNA fragmentation and advanced chromatin condensation. The other pathway, which is caspase-independent, involves AIF and leads to large-scale DNA fragmentation and peripheral chromatin condensation.


Asunto(s)
Apoptosis/fisiología , Caspasas/metabolismo , Núcleo Celular/metabolismo , Flavoproteínas/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Proteínas/metabolismo , Animales , Antineoplásicos/farmacología , Factor Inductor de la Apoptosis , Factor Apoptótico 1 Activador de Proteasas , Arsenitos/farmacología , Caspasa 3 , Caspasas/genética , Células Cultivadas , Cisplatino/farmacología , Grupo Citocromo c/genética , Grupo Citocromo c/metabolismo , Fragmentación del ADN , Desoxirribonucleasas/genética , Desoxirribonucleasas/metabolismo , Etopósido/farmacología , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Flavoproteínas/genética , Colorantes Fluorescentes/metabolismo , Células HeLa , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Microinyecciones , Proteínas/genética , Proteínas Recombinantes/metabolismo
7.
FEBS Lett ; 476(3): 118-23, 2000 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-10913597

RESUMEN

Apoptosis-inducing factor (AIF) is encoded by one single gene located on the X chromosome. AIF is ubiquitously expressed, both in normal tissues and in a variety of cancer cell lines. The AIF precursor is synthesized in the cytosol and is imported into mitochondria. The mature AIF protein, a flavoprotein (prosthetic group: flavine adenine dinucleotide) with significant homology to plant ascorbate reductases and bacterial NADH oxidases, is normally confined to the mitochondrial intermembrane space. In a variety of different apoptosis-inducing conditions, AIF translocates through the outer mitochondrial membrane to the cytosol and to the nucleus. Ectopic (extra-mitochondrial) AIF induces nuclear chromatin condensation, as well as large scale ( approximately 50 kb) DNA fragmentation. Thus, similar to cytochrome c, AIF is a phylogenetically old, bifunctional protein with an electron acceptor/donor (oxidoreductase) function and a second apoptogenic function. In contrast to cytochrome c, however, AIF acts in a caspase-independent fashion. The molecular mechanisms via which AIF induces apoptosis are discussed.


Asunto(s)
Apoptosis/fisiología , Flavoproteínas/metabolismo , Proteínas de la Membrana/metabolismo , Mitocondrias/metabolismo , Oxidorreductasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Factor Inductor de la Apoptosis , ADN Complementario/genética , Flavoproteínas/genética , Flavoproteínas/farmacología , Expresión Génica , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/farmacología , Oxidorreductasas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Distribución Tisular , Cromosoma X/genética
8.
Cell Death Differ ; 7(4): 368-73, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10773821

RESUMEN

Caspase activation may occur in a direct fashion as a result of CD95 death receptor crosslinking (exogenous pathway) or may be triggered indirectly, via a Bcl-2 inhibitable mitochondrial permeabilization event (endogenous pathway). Thymocyte apoptosis is generally accompanied by proteasome activation. If death is induced by DNA damage, inactivation of p53, overexpression of a Bcl-2 transgene, inhibition of protein synthesis, and antioxidants (N-acetylcyteine, catalase) prevent proteasome activation. Glucocorticoid-induced proteasome activation follows a similar pattern of inhibition except for p53. Caspase inhibition fails to affect proteasome activation induced by topoisomerase inhibition or glucocorticoid receptor ligation. In contrast, caspase activation (but not p53 knockout or Bcl-2 overexpression) does interfere with proteasome activation induced by CD95. Specific inhibition of proteasomes with lactacystin or MG123 blocks caspase activation at a pre-mitochondrial level if thymocyte apoptosis is induced by DNA damage or glucocorticoids. In strict contrast, proteasome inhibition has no inhibitory effect on the mitochondrial and nuclear phases of apoptosis induced via CD95. Thus, proteasome activation is a critical event of thymocyte apoptosis stimulated via the endogenous pathway yet dispensable for CD95-triggered death.


Asunto(s)
Apoptosis/fisiología , Cisteína Endopeptidasas/metabolismo , Complejos Multienzimáticos/metabolismo , Linfocitos T/fisiología , Receptor fas/fisiología , Acetilcisteína/farmacología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Inhibidores de Caspasas , Caspasas/metabolismo , Catalasa/farmacología , Células Cultivadas , Cisteína Endopeptidasas/efectos de los fármacos , Dactinomicina/farmacología , Dexametasona/farmacología , Activación Enzimática , Etopósido/farmacología , Femenino , Ratones , Ratones Endogámicos BALB C , Mitocondrias/metabolismo , Complejos Multienzimáticos/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal , Linfocitos T/citología , Linfocitos T/efectos de los fármacos
9.
FASEB J ; 14(5): 729-39, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10744629

RESUMEN

Apoptosis inducing factor (AIF) is a novel apoptotic effector protein that induces chromatin condensation and large-scale ( approximately 50 kbp) DNA fragmentation when added to purified nuclei in vitro. Confocal and electron microscopy reveal that, in normal cells, AIF is strictly confined to mitochondria and thus colocalizes with heat shock protein 60 (hsp60). On induction of apoptosis by staurosporin, c-Myc, etoposide, or ceramide, AIF (but not hsp60) translocates to the nucleus. This suggests that only the outer mitochondrial membrane (which retains AIF in the intermembrane space) but not the inner membrane (which retains hsp60 in the matrix) becomes protein permeable. The mitochondrio-nuclear redistribution of AIF is prevented by a Bcl-2 protein specifically targeted to mitochondrial membranes. The pan-caspase inhibitor Z-VAD. fmk does not prevent the staurosporin-induced translocation of AIF, although it does inhibit oligonucleosomal DNA fragmentation and arrests chromatin condensation at an early stage. ATP depletion is sufficient to cause AIF translocation to the nucleus, and this phenomenon is accelerated by the apoptosis inducer staurosporin. However, in conditions in which both glycolytic and respiratory ATP generation is inhibited, cells fail to manifest any sign of chromatin condensation and advanced DNA fragmentation, thus manifesting a 'necrotic' phenotype. Both in the presence of Z-VAD. fmk and in conditions of ATP depletion, AIF translocation correlates with the appearance of large-scale DNA fragmentation. Altogether, these data are compatible with the hypothesis that AIF is a caspase-independent mitochondrial death effector responsible for partial chromatinolysis.


Asunto(s)
Apoptosis/fisiología , Flavoproteínas/metabolismo , Proteínas de la Membrana/metabolismo , Necrosis , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/efectos de los fármacos , Factor Inductor de la Apoptosis , Transporte Biológico Activo , Caspasas/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Cromatina/metabolismo , Grupo Citocromo c/metabolismo , Daño del ADN , Humanos , Microscopía Confocal , Microscopía Electrónica , Mitocondrias/metabolismo , Modelos Biológicos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ratas , Estaurosporina/farmacología
10.
J Exp Med ; 191(1): 33-46, 2000 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-10620603

RESUMEN

Viral protein R (Vpr) encoded by HIV-1 is a facultative inducer of apoptosis. When added to intact cells or purified mitochondria, micromolar and submicromolar doses of synthetic Vpr cause a rapid dissipation of the mitochondrial transmembrane potential (DeltaPsi(m)), as well as the mitochondrial release of apoptogenic proteins such as cytochrome c or apoptosis inducing factor. The same structural motifs relevant for cell killing are responsible for the mitochondriotoxic effects of Vpr. Both mitochondrial and cytotoxic Vpr effects are prevented by Bcl-2, an inhibitor of the permeability transition pore complex (PTPC). Coincubation of purified organelles revealed that nuclear apoptosis is only induced by Vpr when mitochondria are present yet can be abolished by PTPC inhibitors. Vpr favors the permeabilization of artificial membranes containing the purified PTPC or defined PTPC components such as the adenine nucleotide translocator (ANT) combined with Bax. Again, this effect is prevented by addition of recombinant Bcl-2. The Vpr COOH terminus binds purified ANT, as well as a molecular complex containing ANT and the voltage-dependent anion channel (VDAC), another PTPC component. Yeast strains lacking ANT or VDAC are less susceptible to Vpr-induced killing than control cells yet recover Vpr sensitivity when retransfected with yeast ANT or human VDAC. Hence, Vpr induces apoptosis via a direct effect on the mitochondrial PTPC.


Asunto(s)
Apoptosis , Productos del Gen vpr/fisiología , VIH-1/fisiología , Mitocondrias/fisiología , Sistema Libre de Células , Productos del Gen vpr/química , Humanos , Células Jurkat , Permeabilidad , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana
11.
J Am Soc Nephrol ; 10(3): 507-18, 1999 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10073601

RESUMEN

Even 10 yr after the identification of the antiphospholipid syndrome (APS), renal involvement in the course of APS is still relatively unrecognized, and is probably underestimated. The association of anticardiolipin antibodies and/or lupus anticoagulant with the development of a vaso-occlusive process involving numerous organs is now confirmed. In a multicenter study, 16 cases of "primary" APS (PAPS) were found and followed for 5 yr or more, all with renal biopsy. In all 16 cases of PAPS, there was a vascular nephropathy characterized by small vessel vaso-occlusive lesions associated with fibrous intimal hyperplasia of interlobular arteries (12 patients), recanalizing thrombi in arteries and arterioles (six patients), and focal cortical atrophy (10 patients). In combination, these led to progressive destruction of the kidney, accelerated by acute glomerular and arteriolar microangiopathy in five patients. Focal cortical atrophy is a distinctive lesion, present in 10 biopsies, and likely represents the histologic and functional renal analogue to the multiple cerebral infarcts detected on imaging studies. The clinical hallmark of this vascular nephropathy in PAPS is systemic hypertension, only variably associated with renal insufficiency, proteinuria, or hematuria. The ensemble of histologic renal lesions defined in this study should aid in the separation of the lesions found in cases of secondary APS, especially systemic lupus erythematosus, into those lesions related to APS and those related to the underlying disease.


Asunto(s)
Anticuerpos Antifosfolípidos/análisis , Síndrome Antifosfolípido/patología , Enfermedades Renales/patología , Obstrucción de la Arteria Renal/patología , Trombosis/patología , Adulto , Síndrome Antifosfolípido/complicaciones , Biopsia con Aguja , Femenino , Humanos , Riñón/irrigación sanguínea , Riñón/patología , Enfermedades Renales/etiología , Enfermedades Renales/inmunología , Masculino , Persona de Mediana Edad , Pronóstico , Obstrucción de la Arteria Renal/etiología , Estudios Retrospectivos , Índice de Severidad de la Enfermedad , Tromboflebitis/etiología , Tromboflebitis/patología , Trombosis/etiología
12.
J Exp Med ; 186(1): 25-37, 1997 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-9206994

RESUMEN

According to current understanding, cytoplasmic events including activation of protease cascades and mitochondrial permeability transition (PT) participate in the control of nuclear apoptosis. However, the relationship between protease activation and PT has remained elusive. When apoptosis is induced by cross-linking of the Fas/APO-1/CD95 receptor, activation of interleukin-1beta converting enzyme (ICE; caspase 1) or ICE-like enzymes precedes the disruption of the mitochondrial inner transmembrane potential (DeltaPsim). In contrast, cytosolic CPP32/ Yama/Apopain/caspase 3 activation, plasma membrane phosphatidyl serine exposure, and nuclear apoptosis only occur in cells in which the DeltaPsim is fully disrupted. Transfection with the cowpox protease inhibitor crmA or culture in the presence of the synthetic ICE-specific inhibitor Ac-YVAD.cmk both prevent the DeltaPsim collapse and subsequent apoptosis. Cytosols from anti-Fas-treated human lymphoma cells accumulate an activity that induces PT in isolated mitochondria in vitro and that is neutralized by crmA or Ac-YVAD.cmk. Recombinant purified ICE suffices to cause isolated mitochondria to undergo PT-like large amplitude swelling and to disrupt their DeltaPsim. In addition, ICE-treated mitochondria release an apoptosis-inducing factor (AIF) that induces apoptotic changes (chromatin condensation and oligonucleosomal DNA fragmentation) in isolated nuclei in vitro. AIF is a protease (or protease activator) that can be inhibited by the broad spectrum apoptosis inhibitor Z-VAD.fmk and that causes the proteolytical activation of CPP32. Although Bcl-2 is a highly efficient inhibitor of mitochondrial alterations (large amplitude swelling + DeltaPsim collapse + release of AIF) induced by prooxidants or cytosols from ceramide-treated cells, it has no effect on the ICE-induced mitochondrial PT and AIF release. These data connect a protease activation pathway with the mitochondrial phase of apoptosis regulation. In addition, they provide a plausible explanation of why Bcl-2 fails to interfere with Fas-triggered apoptosis in most cell types, yet prevents ceramide- and prooxidant-induced apoptosis.


Asunto(s)
Apoptosis/inmunología , Ceramidas/farmacología , Cisteína Endopeptidasas/inmunología , Receptor fas/inmunología , Apoptosis/efectos de los fármacos , Caspasa 1 , Cisteína Endopeptidasas/metabolismo , Activación Enzimática , Citometría de Flujo , Humanos , Mitocondrias/inmunología , Mitocondrias/metabolismo , Células Tumorales Cultivadas
13.
J Exp Med ; 184(4): 1331-41, 1996 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-8879205

RESUMEN

Bcl-2 belongs to a family of apoptosis-regulatory proteins which incorporate into the outer mitochondrial as well as nuclear membranes. The mechanism by which the proto-oncogene product Bcl-2 inhibits apoptosis is thus far elusive. We and others have shown previously that the first biochemical alteration detectable in cells undergoing apoptosis, well before nuclear changes become manifest, is a collapse of the mitochondrial inner membrane potential (delta psi m), suggesting the involvement of mitochondrial products in the apoptotic cascade. Here we show that mitochondria contain a pre-formed approximately 50-kD protein which is released upon delta psi m disruption and which, in a cell-free in vitro system, causes isolated nuclei to undergo apoptotic changes such as chromatin condensation and internucleosomal DNA fragmentation. This apoptosis-inducing factor (AIF) is blocked by N-benzyloxycarbonyl-Val-Ala-Asp.fluoromethylketone (Z-VAD.fmk), an antagonist of interleukin-1 beta-converting enzyme (ICE)-like proteases that is also an efficient inhibitor of apoptosis in cells. We have tested the effect of Bcl-2 on the formation, release, and action of AIF. When preventing mitochondrial permeability transition (which accounts for the pre-apoptotic delta psi m disruption in cells), Bcl-2 hyperexpressed in the outer mitochondrial membrane also impedes the release of AIF from isolated mitochondria in vitro. In contrast, Bcl-2 does not affect the formation of AIF, which is contained in comparable quantities in control mitochondria and in mitochondria from Bcl-2-hyperexpressing cells. Furthermore, the presence of Bcl-2 in the nuclear membrane does not interfere with the action of AIF on the nucleus, nor does Bcl-2 hyperexpression protect cells against AIF. It thus appears that Bcl-2 prevents apoptosis by favoring the retention of an apoptogenic protease in mitochondria.


Asunto(s)
Apoptosis/efectos de los fármacos , Endopeptidasas/farmacología , Mitocondrias Hepáticas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/farmacología , Clorometilcetonas de Aminoácidos/farmacología , Animales , Compartimento Celular , Núcleo Celular/efectos de los fármacos , Femenino , Ratones , Ratones Endogámicos BALB C , Modelos Biológicos , Permeabilidad , Inhibidores de Proteasas/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA