Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
2.
Cell Death Differ ; 17(11): 1739-50, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20467439

ABSTRACT

The early-response gene product IEX-1 (also known as IER3) was recently found to interact with the anti-apoptotic Bcl-2 family member, myeloid cell leukemia-1 (Mcl-1). In this study we show that this interaction specifically and timely controls the accumulation of Mcl-1 in the nucleus in response to DNA damage. The IEX-1 protein is rapidly induced by γ-irradiation, genotoxic agents or replication inhibitors, in a way dependent on ataxia telangiectasia mutated (ATM) activity and is necessary for Mcl-1 nuclear translocation. Conversely, IEX-1 protein proteasomal degradation triggers the return of Mcl-1 to the cytosol. IEX-1 and Mcl-1 are integral components of the DNA damage response. Loss of IEX-1 or Mcl-1 leads to genomic instability and increased sensitivity to genotoxic and replicative stresses. The two proteins cooperate to maintain Chk1 activation and G2 checkpoint arrest. Mcl-1 nuclear translocation may foster checkpoint and improve the tumor resistance to DNA damage-based cancer therapies. Deciphering the pathways involved in IEX-1 degradation should lead to the discovery of new therapeutic targets to increase sensitivity of tumor cells to chemotherapy.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , DNA Damage , DNA-Binding Proteins/metabolism , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins , Checkpoint Kinase 1 , Checkpoint Kinase 2 , Genes, bcl-2 , Genomic Instability , Humans , Immediate-Early Proteins/deficiency , Membrane Proteins/metabolism , Mice , Mitochondria/metabolism , Mitosis , Myeloid Cell Leukemia Sequence 1 Protein , Phosphorylation , Protein Kinases/metabolism , Proto-Oncogene Proteins c-bcl-2/deficiency
3.
Cell Death Differ ; 16(2): 298-311, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19023333

ABSTRACT

Promyelomonocytic leukemia (PML) is a prominent oncosuppressor whose inactivation is involved in the pathogenesis of hematological and epithelial cancers. Here, we report that PML aggregated in nuclear bodies in syncytia elicited by the envelope glycoprotein complex (Env) of human immunodeficiency virus-1 (HIV-1) in vitro. PML aggregation occurred after the fusion of nuclei (karyogamy) within syncytia but before the apoptotic program was activated. The aggregation of PML was detectable in syncytia present in the brain or lymph nodes from patients with HIV-1 infection, as well as in a fraction of blood leukocytes, correlating with viral status. Using a range of specific inhibitors of PML (the oncogenic PML/RARalpha fusion product or specific small interfering RNAs), we demonstrated that, in Env-elicited syncytia, PML was required for activating phosphorylation of ataxia telangiectasia mutated (ATM), which colocalized with PML in nuclear bodies, in a molecular complex that also involved topoisomerase IIbeta-binding protein 1. PML knockdown thus inhibited the ATM-dependent DNA damage response that culminates in the activation of p53, p53-dependent transcription of pro-apoptotic genes and cell death. Infection of CD4-expressing cells with HIV-1 also induced syncytial apoptosis, which could be suppressed by inhibiting PML. Altogether, these data indicate that PML activation is a critical early event that participates in the apoptotic demise of HIV-1-elicited syncytia.


Subject(s)
Apoptosis , HIV-1 , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , env Gene Products, Human Immunodeficiency Virus/metabolism , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Giant Cells/virology , HeLa Cells , Humans , Promyelocytic Leukemia Protein , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering , Signal Transduction , Tumor Suppressor Protein p53/metabolism
4.
Psychol Med ; 38(10): 1465-74, 2008 Oct.
Article in English | MEDLINE | ID: mdl-17976250

ABSTRACT

BACKGROUND: This study examined healthcare services used by adults diagnosed with an eating disorder (ED) in a large health maintenance organization in the Pacific Northwest. METHOD: Electronic medical records were used to collect information on all out-patient and in-patient visits and medication dispenses, from 2002 to 2004, for adults aged 18-55 years who received an ED diagnosis during 2003. Healthcare services received the year prior to, and following, the receipt of an ED diagnosis were examined. Cases were matched to five comparison health plan members who had a health plan visit close to the date of the matched case's ED diagnosis. RESULTS: Incidence of EDs (0.32% of the 104,130 females, and 0.02% of the 93,628 males) was consistent with prior research employing treatment-based databases, though less than community-based samples. Most cases (50%) were first identified during a primary-care visit and psychiatric co-morbidity was high. Health services use was significantly elevated in all service sectors among those with an ED when compared with matched controls both in the year preceding and that following the receipt of the incident ED diagnosis. Contrary to expectations, healthcare utilization was found to be similarly high across the spectrum of EDs (anorexia nervosa, bulimia nervosa, and eating disorders not otherwise specified). CONCLUSIONS: The elevation in health service use among women both before and after diagnosis suggests that EDs merit identification and treatment efforts commensurate with other mental health disorders (e.g. depression) which have similar healthcare impact.


Subject(s)
Feeding and Eating Disorders/therapy , Health Services/statistics & numerical data , Adolescent , Adult , Anxiety Disorders/epidemiology , Anxiety Disorders/therapy , Comorbidity , Depressive Disorder/epidemiology , Depressive Disorder/therapy , Feeding and Eating Disorders/epidemiology , Female , Humans , Male , Middle Aged , Northwestern United States/epidemiology , Office Visits/statistics & numerical data , Primary Health Care/statistics & numerical data , Stress Disorders, Post-Traumatic/epidemiology , Stress Disorders, Post-Traumatic/therapy
5.
Oncogene ; 27(5): 565-73, 2008 Jan 24.
Article in English | MEDLINE | ID: mdl-17891185

ABSTRACT

We have identified a gene-profile signature for human primary malignant melanoma associated with metastasis to distant sites and poor prognosis. We analyse the differential gene expression by looking at whole biological pathways rather than individual genes. Among the most significant pathways associated with progression to metastasis, we found the DNA replication (P=10(-14)) and the DNA repair pathways (P=10(-16)). We concentrated our analysis on DNA repair and found that 48 genes of this category, among a list of 234 genes, are associated with metastatic progression. These genes belong essentially to the pathways allowing recovery of stalled replication forks due to spontaneous blockage or induced DNA lesions. Because almost all these differentially expressed repair genes were overexpressed in primary tumors with bad prognosis, we speculate that primary melanoma cells that will metastasize try to replicate in a fast and error-free mode. In contrast to the progression from melanocytes to primary melanoma, genetic stability appears to be necessary for a melanoma cell to give rise to distant metastasis. This overexpression of repair genes explains nicely the extraordinary resistance of metastatic melanoma to chemo- and radio-therapy. Our results may open a new avenue for the discovery of drugs active on human metastatic melanoma.


Subject(s)
DNA Repair , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Melanoma/pathology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Cell Transformation, Neoplastic , Gene Expression Profiling , Humans
6.
Pathol Biol (Paris) ; 55(1): 19-28, 2007 Feb.
Article in French | MEDLINE | ID: mdl-16904272

ABSTRACT

Fanconi anemia (FA) is a recessive human cancer prone syndrome featuring bone marrow failure, developmental abnormalities and hypersensitivity to DNA crosslinking agents exposure. 11 among 12 FA gene have been isolated. The biochemical functions of the FANC proteins remain poorly understood. Anyhow, to cope with DNA crosslinks a cell needs a functional FANC pathway. Moreover, the FANC proteins appear to be involved in cell protection against oxidative damage and in the control of TNF-alpha activity. In this review, we describe the current understanding of the FANC pathway and we present how it may be integrated in the complex networks of proteins involved in maintaining the cellular homeostasis.


Subject(s)
Fanconi Anemia Complementation Group Proteins/genetics , Fanconi Anemia/pathology , Bone Marrow Transplantation , Chromosomes, Human/drug effects , Cross-Linking Reagents/adverse effects , DNA/drug effects , DNA Damage , DNA Repair/genetics , Fanconi Anemia/diagnosis , Fanconi Anemia/genetics , Fanconi Anemia/therapy , Fanconi Anemia Complementation Group Proteins/deficiency , Fanconi Anemia Complementation Group Proteins/physiology , Genes, Recessive , Genetic Complementation Test , Genetic Heterogeneity , Genetic Therapy , Hematopoiesis , Homeostasis , Humans , Mutagens/adverse effects , Oxidative Stress , Phenotype , Tumor Necrosis Factor-alpha/physiology
7.
SAR QSAR Environ Res ; 17(6): 533-47, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17162385

ABSTRACT

The AM1 semiempirical method was employed to study megazol and 13 of its analogues where their activity against Trypanosoma equiperdum was obtained from in vitro tests. Several molecular properties (descriptors or variables) were calculated for the 14 compounds studied to be correlated with the biological activity. For a practical analysis of large data sets, it is necessary to reduce the dimensionality and select the most relevant descriptors related to the biological activity under study. For this purpose, the following chemometric methods were employed: principal component analysis (PCA), hierarchical cluster analysis (HCA), K-nearest neighbour (KNN), stepwise discriminant analysis (SDA) and soft independent modelling of class analogy (SIMCA). These methods showed that the descriptors molecular electronic energy (Eelet), charge on the first nitrogen at substituent 2 (qN), volume of substituent at C5 position (V-S5), dihedral angle (D3) and bond length between atom C4 and its substituents (L4) are responsible for the separation between active and inactive compounds against T. equiperdum.


Subject(s)
Thiadiazoles/chemistry , Thiadiazoles/pharmacology , Trypanosoma/metabolism , Trypanosomiasis/drug therapy , Animals , Chemistry, Pharmaceutical/methods , Cluster Analysis , Drug Design , Models, Chemical , Molecular Structure , Principal Component Analysis , Structure-Activity Relationship
8.
Exp Cell Res ; 269(1): 2-12, 2001 Sep 10.
Article in English | MEDLINE | ID: mdl-11525634

ABSTRACT

Caspase-8 plays an essential role in apoptosis induced by Fas activation. Moreover, caspase-8 can be processed also in response to exposure to genotoxic agents. To decipher the role of caspase-8 in DNA damaging agent (DDA)-induced apoptosis as well as the pathway(s) leading to its activation in response to genotoxic stress, we investigated caspase-8 processing induced by ionizing radiation (IR) or mitomycin C (MMC) treatment in human B-lymphoblasts. Altogether, our observations establish that caspase-8 is actively processed in both receptor-mediated and DDA-induced cell death. However, while Fas-dependent apoptosis absolutely required caspase-8 activity, it is not necessary for completion of the apoptotic program induced by IR and MMC. Experiments performed to understand the molecular pathway(s) of the caspase-8 activation after DDA demonstrated that for both IR and MMC, the Fas/Fas-L interaction is dispensable. Data obtained from caspase inhibitors and from lymphoblasts carrying mutations in ATM and FANCC proteins, involved in DDA response, clearly showed that distinct mechanisms are responsible for caspase-8 activation by IR and MMC in B-lymphoblasts. IR-dependent processing of caspase-8 involves ATM, mitochondrial collapse, FANCC, and caspase-3 activation. Caspase-8 activation by MMC evokes the mitochondrial pathways involving FANCC but not ATM. Collectively, our data indicate that caspase-8 activation is essentially a bystander effect and not a major determinant of the behavior of DDA-exposed cells.


Subject(s)
Apoptosis/physiology , B-Lymphocytes/metabolism , Caspases/metabolism , Cell Cycle Proteins , DNA Damage/physiology , DNA-Binding Proteins , Mutagens/pharmacology , Myeloid Progenitor Cells/metabolism , Nuclear Proteins , fas Receptor/metabolism , Apoptosis/drug effects , Apoptosis/radiation effects , Ataxia Telangiectasia Mutated Proteins , B-Lymphocytes/drug effects , B-Lymphocytes/radiation effects , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/drug effects , Caspases/radiation effects , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Cells, Cultured/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , Enzyme Inhibitors/pharmacology , Fanconi Anemia Complementation Group C Protein , Fanconi Anemia Complementation Group Proteins , Fas Ligand Protein , Humans , Immunohistochemistry , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/metabolism , Mitomycin/pharmacology , Myeloid Progenitor Cells/drug effects , Myeloid Progenitor Cells/radiation effects , Nucleic Acid Synthesis Inhibitors/pharmacology , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/radiation effects , Proteins/drug effects , Proteins/metabolism , Proteins/radiation effects , Signal Transduction/drug effects , Signal Transduction/physiology , Signal Transduction/radiation effects , Tumor Suppressor Proteins , fas Receptor/drug effects , fas Receptor/radiation effects
9.
Int J Radiat Biol ; 76(6): 841-51, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10902739

ABSTRACT

PURPOSE: To define the role of the ataxia telangiectasia (A-T) mutated gene (ATM) in activation and progress of apoptosis. MATERIAL AND METHODS: Three normal and three A-T EBV-transformed cell lines were studied. Following irradiation (IR), Fas activation or ceramide exposure, viability and apoptosis were measured by trypan blue dye exclusion assay and as sub-G1 cell fraction by flow cytometric analysis of propidium iodide stained cultures, respectively. Activation of caspase-3 was evaluated by immunoblot and by an in vitro activity assay on cytosolic cell extracts. To assess changes in mitochondrial potential and reactive oxygen species, cells were stained by 3,3'-dihexyloxacarbocynine iodide or hydroethidine, respectively, and scored by flow cytometry. RESULTS: The observations establish that A-T cells are equipped with a proficient apoptotic machinery, as demonstrated by their ability to undergo mitochondrial collapse and caspase-3 activation after Fas activation or ceramide treatment. Both treatments have a similar cytotoxic effect on normal and A-T cells. In contrast, in spite of the stronger cytotoxicity induced by IR exposure, irradiated A-T cells are unable to undergo mitochondrial collapse and caspase-3 activation. CONCLUSIONS: The data indicate that ATM is necessary in the initiation of molecular pathway(s) leading to IR-induced apoptosis, and suggest that increased radiosensitivity of A-T cells is more likely a direct consequence of necrotic cell death.


Subject(s)
Apoptosis/physiology , Apoptosis/radiation effects , Ataxia Telangiectasia/metabolism , Mitochondria/metabolism , Mitochondria/radiation effects , Protein Serine-Threonine Kinases/metabolism , Apoptosis/genetics , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/pathology , Ataxia Telangiectasia Mutated Proteins , Caspase 3 , Caspases/metabolism , Cell Cycle Proteins , Cell Line , Cell Line, Transformed , DNA-Binding Proteins , Enzyme Activation/radiation effects , Humans , Poly(ADP-ribose) Polymerases/metabolism , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins
10.
Biochimie ; 82(1): 51-8, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10717387

ABSTRACT

Fanconi anemia (FA) is a human genetic disease featuring cancer predisposition, genetic instability and DNA damage hypersensitivity. Although abnormalities in DNA repair and cell cycle checkpoint have been proposed as the underlying defect in this syndrome, these hypotheses did not provide full explanations of the complex phenotype. Although not exclusive of such possibilities, alterations in the control of apoptosis might account for the pleiotropic phenotype of this syndrome. We and others have previously reported a deregulation of the apoptotic response to mitomycin C, suggesting that the products of the Fanconi anemia group C protein (FANCC) contribute to the regulation of apoptosis. To explore the functional importance of the apoptotic alterations in FA we analyzed biochemical steps of the execution phase of apoptosis stimulated by another DNA damaging agent, the gamma-ray using FA cell lines derived from complementation group C (FA-C) independent patients. It is shown that the poly(ADP-ribose) polymerase, a target of caspase-3, is not cleaved in FA-C after ionizing radiation (IR). Moreover, caspase-3 is not processed in its active form and, its activity is not increased by IR in FA-C cells compared to normal cells. Altogether, these results demonstrate that loss of the FANCC activity results in a deficiency of the IR-induced apoptosis which is due to an inability to activate caspase-3. Our work suggests that apoptosis signaling induced by mitomycin C and IR is subject to common regulation involving the FANCC protein.


Subject(s)
Caspases/metabolism , Cell Cycle Proteins , DNA-Binding Proteins , Fanconi Anemia/metabolism , Fanconi Anemia/radiotherapy , Nuclear Proteins , Proteins/metabolism , Apoptosis/radiation effects , Caspase 3 , Caspase Inhibitors , Caspases/radiation effects , Cell Death/radiation effects , Cell Line , Coumarins/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Activation/radiation effects , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group C Protein , Fanconi Anemia Complementation Group Proteins , Gamma Rays , Humans , Oligopeptides/metabolism , Oligopeptides/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Poly(ADP-ribose) Polymerases/radiation effects , Proteins/genetics , Proteins/radiation effects , Radiation, Ionizing
11.
Exp Cell Res ; 246(2): 384-94, 1999 Feb 01.
Article in English | MEDLINE | ID: mdl-9925754

ABSTRACT

Deregulation of apoptosis seems to be a hallmark of the Fanconi anemia (FA) syndrome. In order to further define the role of the FA protein from complementation group C (FAC) in apoptosis, we characterized parameters modified during the mitomycin-C (MMC)-induced apoptotic program. It is shown that despite a higher level of cell death for FA compared to normal lymphoblasts after MMC treatment, FA cells do not display a marked DNA fragmentation. Furthermore, while playing a central role in MMC apoptosis of normal lymphoblasts, the activity of caspase-3-like proteases is altered in FA cells. Interestingly, the disruption of the mitochondrial transmembrane potential (Deltapsi), an early event that can lead to apoptotic or to necrotic death, is accomplished similarly in FA and in normal cells. Finally, it is shown that the overexpressed FAC protein inhibited the apoptotic steps, with the exception of the decrease of the Deltapsi. Altogether, our results indicate that the FAC protein acts at a step preceding the activation of the caspases and after the modification of the Deltapsi, a decision point at which cells can be pushed toward either apoptosis or necrosis and which, consequently, regulates the balance between the two modes of cell death.


Subject(s)
Apoptosis , Cell Cycle Proteins , DNA-Binding Proteins , Necrosis , Nuclear Proteins , Proteins/physiology , Caspase 3 , Caspases/metabolism , Caspases/physiology , DNA Fragmentation , Enzyme Activation , Fanconi Anemia Complementation Group C Protein , Fanconi Anemia Complementation Group Proteins , Gene Expression Regulation , Humans , Membrane Potentials , Mitochondria , Mitomycin/pharmacology , Nucleosomes , Proteins/genetics , Tumor Cells, Cultured
12.
Int J Oncol ; 13(1): 107-14, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9625811

ABSTRACT

Wild-type (wt) tumor suppressor p53 has been implicated in cellular radiosensitivity, mediated by its role in apoptosis and growth arrest. Intriguingly, it was observed that the temperature sensitive (ts) mutant p53val135 protein functions as a positive modulator of cellular radiosensitivity, as evident from acceleration of irradiation-induced apoptosis of M1p53ts (p53val135) cells at the non-permissive temperature; this effect was correlated with acceleration of exit from the G2 checkpoint of the cell cycle. In this work it is shown that the ability of mutant p53val135 to accelerate irradiation-induced apoptosis, at the non-permissive temperature, was devoid of transcriptional trans-activation of p53 target genes. In contrast, the apoptotic function of wt p53val135 was observed to include components which are both dependent and independent of transcriptional trans-activation. Taken together, these observations suggest that mutant p53val135 protein retains the apoptotic component of wt p53 that is devoid of transcriptional trans-activation, and that, although this activity is insufficient to induce apoptosis on its own, it can cooperate to accelerate DNA damage-induced cell death. The results of this work contribute to a better understanding of the complexity of the apoptotic response elicited by wt p53, and highlight the potential role of mutant p53 proteins, as well as trans-activation independent apoptosis, in tumor suppression by irradiation therapy.


Subject(s)
Apoptosis/genetics , Transcriptional Activation , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis/drug effects , Cycloheximide/pharmacology , DNA Damage/drug effects , Mice , Mutagenesis , Protein Synthesis Inhibitors/pharmacology , Tumor Cells, Cultured
13.
Int J Radiat Oncol Biol Phys ; 40(5): 1019-26, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9539555

ABSTRACT

PURPOSE: Patients with a history of head and neck irradiation in childhood are at risk to develop thyroid tumors. The aim of this study was to determine if an impairement of DNA strand breaks repair could account for this observation. METHODS AND MATERIALS: Circulating unstimulated lymphocytes of a group of 13 patients who developed thyroid tumors after radiotherapy were submitted to the alkaline single-cell gel electrophoresis assay (SCGE or "comet" assay) after in vitro exposure to 2 and 5 Gy of gamma-rays. A control group of 8 healthy donors and 2 cases with a history of neck irradiation who did not develop a thyroid tumor were also analysed. The immediate response was compared to that observed after 15, 30, and 60 min of postexposure incubation periods. RESULTS: Induction of DNA strand breaks is a dose-dependent process. The SCGE assay parameters did not differ significantly between patients and controls immediately (t=0) after irradiation at the two doses used. As compared to healthy donors, a slower kinetics of repair was found in the patients. The proportion of residual damage at 60 min postirradiation was significantly (p < 0.01) higher in patients than in controls, at both doses analysed. Flow cytometric analysis of apoptosis and p53 protein status studied before and after irradiation showed no apparent relationship with the repair capacity. CONCLUSION: This preliminary study suggests that a subgroup of patients who develop thyroid tumors after a history of irradiation are partially defective in the late restitution of in vitro radiation-induced DNA strand breaks. This deficiency could be a predisposing factor to radiation-associated thyroid tumorigenesis. Detection of susceptible individuals using the simple and rapid comet assay, especially children receiving radiotherapeutic treatment, may allow a preventive surveillance for radiation-associated epithelial thyroid tumor development.


Subject(s)
DNA Repair , Neoplasms, Radiation-Induced/genetics , Thyroid Neoplasms/genetics , Adult , Apoptosis , DNA/radiation effects , DNA Damage , Electrophoresis/methods , Female , Humans , Male , Middle Aged , Thyroid Gland/radiation effects
14.
Eur Heart J ; 19 Suppl B: B25-35, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9519349

ABSTRACT

Increased mortality and reduced functional capacity are the two main characteristics of chronic heart failure. Activation of the renin-angiotensin and sympathetic systems has a primary role in the progressive worsening of heart failure and increased mortality of patients. In addition, both systems may be important in the pathogenesis of exercise intolerance, although there is only a weak relationship between neurohormonal activation and exercise capacity. While neurohormonal antagonists, such as angiotensin-converting enzyme (ACE) inhibitors and beta-blockers, consistently improve the prognosis of patients with heart failure, their effects on exercise tolerance have often been less significant. This problem has been emphasized by the introduction of beta-blockers for the therapy of heart failure. Beta blockade results in a significant improvement in left ventricular function during rest and exercise. However, the reduction in chronotropic response to exercise as well as the metabolic changes caused by these agents in skeletal muscle may result in an apparent lack of change in maximal functional capacity. This effect is particularly important with the new third generation non-selective beta-blockers. The pronounced anti-adrenergic activity of these compounds accounts for their greater negative chronotropic effect and relates to the lack of improvement in peak oxygen consumption (VO2). Submaximal exercise testing can be used to assess changes induced by these agents. However, even the six-minute walk test may act as an almost maximal test in patients with advanced heart failure: moreover, the measurement of submaximal exercise duration may be sensitive enough to detect changes in single-centre trials, but not in multicentre trials. To date, direct assessment of symptoms by both patient and physician is still the most sensitive tool to monitor changes in functional status with non-selective beta-blockers. Thus, an accurate method of measuring patients' symptoms, in addition to the clinical examination, is still necessary when neurohormonal antagonists are used in patients with chronic heart failure.


Subject(s)
Cardiac Output, Low/drug therapy , Cardiac Output, Low/physiopathology , Neurotransmitter Agents/antagonists & inhibitors , Quality of Life , Adrenergic beta-Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Humans , Physical Exertion
15.
Apoptosis ; 3(4): 229-36, 1998 Sep.
Article in English | MEDLINE | ID: mdl-14646486

ABSTRACT

Fanconi anemia (FA) is a rare recessive, human genetic syndrome characterized by progressive bone marrow failure, developmental abnormalities, predisposition to malignancy, chromosomal instability and DNA damage hypersensitivity. Two (FAA and FAC) of the five genes involved were cloned but their functions remain unknown. At present, the involvement of FA proteins in DNA repair, redox status of the cell and apoptosis are areas of intensive investigation. The aim of this review is to synthesize current results and ideas concerning the involvement of apoptosis in the FA phenotype and conversely, the role of FA proteins in the control of apoptosis.

16.
Cardiologia ; 42(5): 503-12, 1997 May.
Article in Italian | MEDLINE | ID: mdl-9289367

ABSTRACT

Carvedilol has been shown to determine a significant improvement in left ventricular function, symptoms, clinical course and prognosis of patients with chronic heart failure. However, these results were obtained in medium-term studies of < 1 year duration. We report the results obtained with long-term (3-4 years) carvedilol administration to 40 patients with idiopathic dilated cardiomyopathy who were initially recruited in a 4-month double-blind placebo-controlled trial. In the initial 4-month double-blind trial, 20 patients were randomized to placebo and 20 to carvedilol treatment. All patients, except one who was not on ACE-inhibitors, were on digoxin, furosemide and ACE-inhibitors. Carvedilol or placebo doses were progressively titrated, at weekly intervals, up to the maximal doses of 25 mg bid. After the initial 4-month double-blind phase, all patients were followed long term. Mean follow-up duration was 52 +/- 12 months (range 48-61). Among the 20 patients initially randomized to carvedilol administration, 4 died (3 for cardiac and 1 for extracardiac causes) and 2 underwent heart transplant. Among the 20 patients initially randomized to placebo, 5 died for cardiac causes, 3 underwent heart transplant and 4 were started on carvedilol because of progressive heart failure during the initial 4 months of the study. The remaining 8 patients, who were kept on digoxin, furosemide and ACE-inhibitors, were used as control group. Each patient underwent an assessment of clinical conditions (NYHA functional classification and Minnesota Living with Heart Failure questionnaire), equilibrium radionuclide ventriculography, and maximal cardiopulmonary bicycle exercise testing. Exams were performed before treatment, after 4 and 12 months, and at the end of the follow-up period. No significant difference between the carvedilol and control group was present at baseline. Compared with baseline, patients in the control group presented a significant increase in left ventricular end-diastolic volume after long-term follow-up (from 126 +/- 62 to 138 +/- 43 and 158 +/- 52 ml/m2 after 12 and 48 months, respectively). No significant difference, compared to baseline values, was noted. Patients on carvedilol presented a persistent improvement in left ventricular function. This was shown by the progressive increment in left ventricular ejection fraction from 22 +/- 6 to 34 +/- 11, 37 +/- 11 and 37 +/- 13%, after 4, 12 and 48 months, respectively (p < 0.001) with a concomitant reduction in left ventricular end-diastolic volume from 147 +/- 54 to 101 +/- 44 ml/m2 at the end of the follow-up (p < 0.05). NYHA functional class remained significantly improved, in comparison with baseline (2.6 +/- 0.5 to 1.9 +/- 0.3, 1.9 +/- 0.8 and 2.0 +/- 1.0 after 4, 12 and 48 months, respectively; p < 0.01). Maximal functional capacity, assessed as peak VO2 was not significantly changed after 4 months (from 15.2 +/- 3.6 to 16.4 +/- 4.0 ml/kg/min) and showed a tendency towards a further improvement after 12 months and at the end of the follow-up (17.3 +/- 5.6 and 17.2 +/- 5.3 ml/kg/min, respectively). These results show that the favorable effects of carvedilol administration on left ventricular function and clinical symptoms are maintained also after long-term treatment.


Subject(s)
Adrenergic beta-Antagonists/therapeutic use , Carbazoles/therapeutic use , Cardiomyopathy, Dilated/drug therapy , Propanolamines/therapeutic use , Adrenergic beta-Antagonists/administration & dosage , Adrenergic beta-Antagonists/adverse effects , Carbazoles/administration & dosage , Carbazoles/adverse effects , Carvedilol , Double-Blind Method , Female , Follow-Up Studies , Humans , Male , Middle Aged , Propanolamines/administration & dosage , Propanolamines/adverse effects
17.
Cancer Res ; 57(9): 1722-30, 1997 May 01.
Article in English | MEDLINE | ID: mdl-9135015

ABSTRACT

Fanconi anemia (FA) is a genetic human disorder associated with bone marrow failure and predisposition to cancer. FA cells show poor growth capacity and spontaneous chromosomal anomalies as well as cellular and chromosomal hypersensitivity to DNA cross-linking agents such as mitomycin C (MMC). Because it is likely that disruption of the apoptotic control would lead to such a phenotype, we investigated the implication of apoptosis in the FA syndrome. It is shown that, although demonstrating a high frequency of spontaneous apoptosis, FA cells from four genetic complementation groups are deficient in gamma-ray-induced apoptosis and their MMC hypersensitivity is not due to apoptosis. Fas is a cell surface receptor belonging to the tumor necrosis factor receptor family and is involved in apoptosis. We show that, independently of DNA damage, the alteration in the control of apoptosis in FA concerns also the pathway initiated by Fas activation. Finally, ectopic expression of the wild-type FAC gene corrects the MMC hypersensitivity and anomalies in apoptosis and cell cycle response in FA cells. Altogether, these findings strongly implicate the FA genes as playing a major role in the control of apoptosis. Thus, further studies with FA syndrome will be instrumental toward molecularly dissecting the apoptotic pathways.


Subject(s)
Apoptosis , Cell Cycle Proteins , DNA-Binding Proteins , Fanconi Anemia/pathology , Nuclear Proteins , Proteins/physiology , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Cycle , Cells, Cultured , DNA Fragmentation , Fanconi Anemia Complementation Group Proteins , Gamma Rays , Genes , Humans , Lymphocytes/radiation effects , Mitomycin/pharmacology , fas Receptor/metabolism
18.
Cancer Res ; 56(6): 1400-4, 1996 Mar 15.
Article in English | MEDLINE | ID: mdl-8640831

ABSTRACT

Ataxia telangiectasia (AT) is a recessive genetic disease featuring neurodegeneration, immunodeficiency, chromosomal instability, radiation hypersensitivity, and increased predisposition to cancer. Reduced or delayed induction of the tumor suppressor protein p53 after gamma -irradiation was reported. These characteristics may be compatible with an inability to correctly regulate apoptosis. We show here that AT lymphocytes and EBV-transformed lymphoblasts demonstrate a significantly higher level of spontaneous apoptosis, whereas ionizing radiation-induced apoptosis is reduced compared to normal cells. However, neither AT nor normal primary fibroblasts undergo apoptosis after irradiation. Consequently, we conclude that the radiosensitivity of the AT cells is not related to an increased apoptotic response. Finally, we show that SV40-transformed AT fibroblasts undergo gamma- ray-induced apoptosis, while SV40-transformed normal cells do not. This result raises the question of the physiological relevance of the latter cellular model with respect to the AT phenotype.


Subject(s)
Apoptosis/physiology , Ataxia Telangiectasia/pathology , Fibroblasts/pathology , Lymphocytes/pathology , Ataxia Telangiectasia/genetics , Cell Line , Cell Line, Transformed , Family , Fibroblasts/radiation effects , Herpesvirus 4, Human , Humans , Lymphocytes/radiation effects , Time Factors
19.
Oncogene ; 10(11): 2263-70, 1995 Jun 01.
Article in English | MEDLINE | ID: mdl-7784074

ABSTRACT

DNA damage in proliferating mammalian cells induces a complex cellular response comprising perturbation of the cell cycle and programmed cell death. The relationship between p53-dependent and p53-independent apoptotic cell death, as well as the cell cycle checkpoints induced by DNA damaging agents were explored in hematopoietic cells, using M1 myeloblastic leukemia cells, which are null for p53 expression, genetically engineered M1 variants, expressing p53ts and bcl-2 transgenes, as well as myeloblast enriched bone-marrow cells obtained from wild type p53 (wt p53) and p53-deficient mice. It is shown that gamma-irradiation of M1p53ts cells activated a function of the temperature sensitive mutant transgene p53 (p53ts), promoting increased apoptosis relative to parental, null p53 M1 cells. It is also shown that the kinetics of apoptotic cell death induced by gamma-irradiation correlated with the rapidity of exit from gamma-ray-induced G2 arrest for all the different hematopoietic cell types indicated above. Finally, data has been obtained to demonstrate that, in addition to a role in apoptosis and G1 arrest, wild-type p53 positively modulated the exit from the gamma-ray-induced G2 checkpoint. Taken together, these findings indicate that this new function for p53 is a component of the physiological pathway by which p53 exerts its role in apoptosis.


Subject(s)
Apoptosis/genetics , DNA Damage , G2 Phase , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis/radiation effects , Gamma Rays , Hot Temperature , Mice , Mutation , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
20.
Oncogene ; 10(1): 9-17, 1995 Jan 05.
Article in English | MEDLINE | ID: mdl-7824283

ABSTRACT

Fanconi anemia belongs to a group of human genetic diseases characterized by chromosomal instability, sensitivity to genotoxic agents associated to impaired processing of DNA lesions, cell cycle anomalies and cancer predisposition. We recently added to this list of distinctive features reduced production of interleukin 6 and overproduction of tumor necrosis factor alpha. Since growth factor deprivation, TNF alpha treatment or DNA damage can trigger apoptosis, we monitored the apoptotic response of FA cell lines. We show here that, although the spontaneous rate of apoptosis is slightly more elevated in FA than in normal cell cultures, the apoptosis induced by gamma-irradiation is drastically reduced in FA. Since the induction of apoptosis by radiation is a p53-dependent mechanism, the induction of this protein in FA cells was also examined. We found that the p53 protein is not radio-induced in FA cells belonging to the two genetic complementation groups examined (C and D), in contrast to normal cells. Moreover, the same impairment in p53 induction is observed after exposure to mitomycin C, a chemical agent for which FA cells demonstrate a specific cellular and chromosomal hypersensitivity, as well as after u.v.-B irradiation, an agent known to cause oxidative stress. These observations are in line with recent reports showing that at least certain cell lines from other chromosome breakage syndromes, such as ataxia telangiectasia and Bloom syndrome, may be also defective for radiation-induced increase of p53 protein. As the p53 tumor suppressor gene encodes a transcriptional activator whose targets include genes that regulate genomic stability, cellular response to DNA damage and cell cycle progression, we suggest that altered expression of p53 may be relevant to the FA phenotype.


Subject(s)
Fanconi Anemia/pathology , Genes, p53 , Lymphocytes/radiation effects , Cell Cycle , Cells, Cultured , Fanconi Anemia/genetics , Humans , Lymphocytes/metabolism , Lymphocytes/pathology , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...