129 results on '"Susin SA"'
Search Results
2. AIF : un nouvel agent double mitochondrial.
- Author
-
Zamzani, N, primary, Susin, SA, additional, and Kroemer, G, additional
- Published
- 1999
- Full Text
- View/download PDF
3. Coopération mortelle entre la protéine pro-apoptotique Bax et le translocateur à adénine nucléotide pour le contrôle mitochondrial de l'apoptose.
- Author
-
Brenner, C, primary, Marzo, I, additional, Zamzami, N, additional, Susin, SA, additional, Vieira, HLA, additional, and Kroemer, G, additional
- Published
- 1998
- Full Text
- View/download PDF
4. The novel retinoid 6-[3-(1-adamantyl)-4-hydroxyphenyl]-2-naphtalene carboxylic acid can trigger apoptosis through a mitochondrial pathway independent of the nucleus
- Author
-
Marchetti, P., Zamzami, N., Bertrand Joseph, Schraen-Maschke, S., Mereau-Richard, C., Costantini, P., Metivier, D., Susin, Sa, Kroemer, G., and Formstecher, P.
- Subjects
Cell Nucleus ,Cytoplasm ,Cell-Free System ,Flavoproteins ,Transcription, Genetic ,Mitochondrial Permeability Transition Pore ,Receptors, Retinoic Acid ,Apoptosis Inducing Factor ,Membrane Proteins ,Antineoplastic Agents ,Apoptosis ,Endoplasmic Reticulum ,Mitochondrial Membrane Transport Proteins ,Ion Channels ,Mitochondria ,Retinoids ,Proto-Oncogene Proteins c-bcl-2 ,Tumor Cells, Cultured ,Humans - Abstract
The novel retinoid 6-[3-(1-adamantyl)-4-hydroxyphenyl]-2-naphtalene carboxylic acid (AHPN/CD437), a retinoic acid receptor (RAR)gamma activator, has been found to inhibit the growth and to induce apoptosis of a wide variety of malignant cell types including solid tumors and various leukemias. Interestingly, CD437 is able to induce apoptosis in some all-trans-retinoic acid (ATRA)-resistant models. In a number of experimental systems, the early apoptotic stage that precedes nuclear chromatinolysis consists in mitochondrial alterations, including a disruption of the inner mitochondrial transmembrane potential (delta(psi)m) mediated by the mitochondrial permeability transition (MPT). Similarly CD437 causes RPMI 8226, a human myeloma cell line, to undergo a rapid delta(psi)m disruption that precedes other apoptotic alterations such as the generation of reactive oxygen species and DNA fragmentation. The same sequence of events is observed during the CD437-induced apoptosis in L363, a RARgamma-negative human myeloma cell line, as well as RPMI 8226 cytoplasts (anucleate cells). Indeed, RPMI 8226 cells and cytoplasts manifest a similar degree in delta(psi)m loss, phosphatidylserine exposure, and caspase activation in response to CD437, which indicates that nuclear effects cannot account for the apoptogenic potential of CD437. The mitochondrial release of cytochrome c, the activation of caspases as well as nuclear signs of CD437-induced apoptosis are fully prevented by the MPT inhibitory compound cyclosporin A. Purified mitochondria can be directly induced to undergo MPT with CD437 but not with ATRA. In a cell-free in vitro system consisting of exposing mitochondrial supernatants to isolated nuclei, only supernatants from CD437-treated mitochondria provoke chromatin condensation, whereas supernatants from mitochondria treated with ATRA, or with the combination of CD437 and cyclosporin A, remain inactive. In conclusion, these results suggest that the rapid execution of CD437-induced apoptosis is a nucleus-independent (and probably RARgamma-independent) phenomenon involving mitochondria and MPT.
5. Epigenetic features support the diagnosis of B-cell prolymphocytic leukemia and identify two clinico-biological subtypes.
- Author
-
Charalampopoulou S, Chapiro E, Nadeu F, Zenz T, Beà S, Aymerich M, Martinez-Farran A, Rozman M, Roos-Weil D, Bernard OA, Susin SA, Parker H, Walewska R, Oakes CC, Strefford JCC, Campo E, Matutes E, Duran-Ferrer M, Nguyen-Khac F, and Martin-Subero JI
- Published
- 2024
- Full Text
- View/download PDF
6. CD38/NAD + glycohydrolase and associated antigens in chronic lymphocytic leukaemia: From interconnected signalling pathways to therapeutic strategies.
- Author
-
Bauvois B, Nguyen-Khac F, Merle-Béral H, and Susin SA
- Abstract
Chronic lymphocytic leukaemia (CLL) is a heterogenous disease characterized by the accumulation of neoplastic CD5
+ /CD19+ B lymphocytes. The spreading of the leukaemia relies on the CLL cell's ability to survive in the blood and migrate to and proliferate within the bone marrow and lymphoid tissues. Some patients with CLL are either refractory to the currently available therapies or relapse after treatment; this emphasizes the need for novel therapeutic strategies that improving clinical responses and overcome drug resistance. CD38 is a marker of a poor prognosis and governs a set of survival, proliferation and migration signals that contribute to the pathophysiology of CLL. The literature data evidence a spatiotemporal association between the cell surface expression of CD38 and that of other CLL antigens, such as the B-cell receptor (BCR), CD19, CD26, CD44, the integrin very late antigen 4 (VLA4), the chemokine receptor CXCR4, the vascular endothelial growth factor receptor-2 (VEGF-R2), and the neutrophil gelatinase-associated lipocalin receptor (NGAL-R). Most of these proteins contribute to CLL cell survival, proliferation and trafficking, and cooperate with CD38 in multilayered signal transduction processes. In general, these antigens have already been validated as therapeutic targets in cancer, and a broad repertoire of specific monoclonal antibodies and derivatives are available. Here, we review the state of the art in this field and examine the therapeutic opportunities for cotargeting CD38 and its partners in CLL, e.g. by designing novel bi-/trispecific antibodies., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024. Published by Elsevier B.V.)- Published
- 2024
- Full Text
- View/download PDF
7. ACOX1-mediated peroxisomal fatty acid oxidation contributes to metabolic reprogramming and survival in chronic lymphocytic leukemia.
- Author
-
Tannoury M, Ayoub M, Dehgane L, Nemazanyy I, Dubois K, Izabelle C, Brousse A, Roos-Weil D, Maloum K, Merle-Béral H, Bauvois B, Saubamea B, Chapiro E, Nguyen-Khac F, Garnier D, and Susin SA
- Subjects
- Humans, B-Lymphocytes metabolism, Fatty Acids metabolism, Fatty Acids therapeutic use, Metabolic Reprogramming, Mitochondria metabolism, Leukemia, Lymphocytic, Chronic, B-Cell pathology
- Abstract
Chronic lymphocytic leukemia (CLL) is still an incurable disease, with many patients developing resistance to conventional and targeted therapies. To better understand the physiology of CLL and facilitate the development of innovative treatment options, we examined specific metabolic features in the tumor CLL B-lymphocytes. We observed metabolic reprogramming, characterized by a high level of mitochondrial oxidative phosphorylation activity, a low glycolytic rate, and the presence of C2- to C6-carnitine end-products revealing an unexpected, essential role for peroxisomal fatty acid beta-oxidation (pFAO). Accordingly, downmodulation of ACOX1 (a rate-limiting pFAO enzyme overexpressed in CLL cells) was enough to shift the CLL cells' metabolism from lipids to a carbon- and amino-acid-based phenotype. Complete blockade of ACOX1 resulted in lipid droplet accumulation and caspase-dependent death in CLL cells, including those from individuals with poor cytogenetic and clinical prognostic factors. In a therapeutic translational approach, ACOX1 inhibition spared non-tumor blood cells from CLL patients but led to the death of circulating, BCR-stimulated CLL B-lymphocytes and CLL B-cells receiving pro-survival stromal signals. Furthermore, a combination of ACOX1 and BTK inhibitors had a synergistic killing effect. Overall, our results highlight a less-studied but essential metabolic pathway in CLL and pave the way towards the development of new, metabolism-based treatment options., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
8. del(8p) and TNFRSF10B loss are associated with a poor prognosis and resistance to fludarabine in chronic lymphocytic leukemia.
- Author
-
Jondreville L, Dehgane L, Doualle C, Smagghe L, Grange B, Davi F, Lerner LK, Garnier D, Bravetti C, Tournilhac O, Roos-Weil D, Boubaya M, Chapiro E, Susin SA, and Nguyen-Khac F
- Subjects
- Humans, Chromosome Aberrations, Mutation, Prognosis, Receptors, TNF-Related Apoptosis-Inducing Ligand genetics, Vidarabine pharmacology, Vidarabine therapeutic use, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy, Leukemia, Lymphocytic, Chronic, B-Cell genetics
- Abstract
Chronic lymphocytic leukemia (CLL) is a heterogeneous disease, the prognosis of which varies according to the cytogenetic group. We characterized a rare chromosomal abnormality (del(8p), deletion of the short arm of chromosome 8) in the context of CLL. By comparing the largest cohort of del(8p) CLL to date (n = 57) with a non-del(8p) cohort (n = 155), del(8p) was significantly associated with a poor prognosis, a shorter time to first treatment, worse overall survival (OS), and a higher risk of Richter transformation. For patients treated with fludarabine-based regimens, the next-treatment-free survival and the OS were shorter in del(8p) cases (including those with mutated IGHV). One copy of the TNFRSF10B gene (coding a pro-apoptotic receptor activated by TRAIL) was lost in 91% of del(8p) CLL. TNFRSF10B was haploinsufficient in del(8p) CLL, and was involved in the modulation of fludarabine-induced cell death - as confirmed by our experiments in primary cells and in CRISPR-edited TNFRSF10B knock-out CLL cell lines. Lastly, del(8p) abrogated the synergy between fludarabine and TRAIL-induced apoptosis. Our results highlight del(8p)'s value as a prognostic marker and suggest that fit CLL patients (i.e. with mutated IGHV and no TP53 disruption) should be screened for del(8p) before the initiation of fludarabine-based treatment., (© 2023. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
9. The Value of Neutrophil Gelatinase-Associated Lipocalin Receptor as a Novel Partner of CD38 in Chronic Lymphocytic Leukemia: From an Adverse Prognostic Factor to a Potential Pharmacological Target?
- Author
-
Bauvois B, Chapiro E, Quiney C, Maloum K, Susin SA, and Nguyen-Khac F
- Abstract
Chronic lymphocytic leukemia (CLL) is characterized by the accumulation of neoplastic B lymphocytes that escape death, and correlates with the expression of negative prognostic markers such as the CD38 antigen. Although certain new drugs approved by the US Food and Drug Administration improve the clinical outcome of CLL patients, drug resistance and disease relapse still occur. Like CD38, neutrophil gelatinase-associated lipocalin receptor (NGAL-R) is frequently overexpressed in CLL cells. Here, we evaluated the concomitant surface expression of NGAL-R and CD38 in leukemic blood cells from 52 CLL patients (37 untreated, 8 in clinical remission, and 7 relapsed). We provide evidence of a positive correlation between NGAL-R and CD38 levels both in the interpatient cohorts ( p < 0.0001) and in individual patients, indicating a constitutive association of NGAL-R and CD38 at the cell level. Patients with progressing CLL showed a time-dependent increase in NGAL-R/CD38 levels. In treated CLL patients who achieved clinical remission, NGAL-R/CD38 levels were decreased, and were significantly lower than in the untreated and relapsed groups ( p < 0.02). As NGAL-R and CD38 participate in CLL cell survival, envisioning their simultaneous inhibition with bispecific NGAL-R/CD38 antibodies capable of inducing leukemic cell death might provide therapeutic benefit for CLL patients.
- Published
- 2023
- Full Text
- View/download PDF
10. Extracellular Vesicles in Chronic Lymphocytic Leukemia: Tumor Microenvironment Messengers as a Basis for New Targeted Therapies?
- Author
-
Dubois K, Tannoury M, Bauvois B, Susin SA, and Garnier D
- Abstract
In addition to intrinsic genomic and nongenomic alterations, tumor progression is also dependent on the tumor microenvironment (TME, mainly composed of the extracellular matrix (ECM), secreted factors, and bystander immune and stromal cells). In chronic lymphocytic leukemia (CLL), B cells have a defect in cell death; contact with the TME in secondary lymphoid organs dramatically increases the B cells' survival via the activation of various molecular pathways, including the B cell receptor and CD40 signaling. Conversely, CLL cells increase the permissiveness of the TME by inducing changes in the ECM, secreted factors, and bystander cells. Recently, the extracellular vesicles (EVs) released into the TME have emerged as key arbiters of cross-talk with tumor cells. The EVs' cargo can contain various bioactive substances (including metabolites, proteins, RNA, and DNA); upon delivery to target cells, these substances can induce intracellular signaling and drive tumor progression. Here, we review recent research on the biology of EVs in CLL. EVs have diagnostic/prognostic significance and clearly influence the clinical outcome of CLL; hence, from the perspective of blocking CLL-TME interactions, EVs are therapeutic targets. The identification of novel EV inhibitors might pave the way to the development of novel combination treatments for CLL and the optimization of currently available treatments (including immunotherapy).
- Published
- 2023
- Full Text
- View/download PDF
11. Current Status of Novel Agents for the Treatment of B Cell Malignancies: What's Coming Next?
- Author
-
Tannoury M, Garnier D, Susin SA, and Bauvois B
- Abstract
Resistance to death is one of the hallmarks of human B cell malignancies and often contributes to the lack of a lasting response to today's commonly used treatments. Drug discovery approaches designed to activate the death machinery have generated a large number of inhibitors of anti-apoptotic proteins from the B-cell lymphoma/leukemia 2 family and the B-cell receptor (BCR) signaling pathway. Orally administered small-molecule inhibitors of Bcl-2 protein and BCR partners (e.g., Bruton's tyrosine kinase and phosphatidylinositol-3 kinase) have already been included (as monotherapies or combination therapies) in the standard of care for selected B cell malignancies. Agonistic monoclonal antibodies and their derivatives (antibody-drug conjugates, antibody-radioisotope conjugates, bispecific T cell engagers, and chimeric antigen receptor-modified T cells) targeting tumor-associated antigens (TAAs, such as CD19, CD20, CD22, and CD38) are indicated for treatment (as monotherapies or combination therapies) of patients with B cell tumors. However, given that some patients are either refractory to current therapies or relapse after treatment, novel therapeutic strategies are needed. Here, we review current strategies for managing B cell malignancies, with a focus on the ongoing clinical development of more effective, selective drugs targeting these molecules, as well as other TAAs and signaling proteins. The observed impact of metabolic reprogramming on B cell pathophysiology highlights the promise of targeting metabolic checkpoints in the treatment of these disorders.
- Published
- 2022
- Full Text
- View/download PDF
12. Keeping Cell Death Alive: An Introduction into the French Cell Death Research Network.
- Author
-
Ichim G, Gibert B, Adriouch S, Brenner C, Davoust N, Desagher S, Devos D, Dokudovskaya S, Dubrez L, Estaquier J, Gillet G, Guénal I, Juin PP, Kroemer G, Legembre P, Levayer R, Manon S, Mehlen P, Meurette O, Micheau O, Mignotte B, Nguyen-Khac F, Popgeorgiev N, Poyet JL, Priault M, Ricci JE, Riquet FB, Susin SA, Suzanne M, Vacher P, Walter L, and Mollereau B
- Subjects
- Animals, Apoptosis, Cell Death, Humans, Necrosis, Caenorhabditis elegans, Neoplasms
- Abstract
Since the Nobel Prize award more than twenty years ago for discovering the core apoptotic pathway in C. elegans , apoptosis and various other forms of regulated cell death have been thoroughly characterized by researchers around the world. Although many aspects of regulated cell death still remain to be elucidated in specific cell subtypes and disease conditions, many predicted that research into cell death was inexorably reaching a plateau. However, this was not the case since the last decade saw a multitude of cell death modalities being described, while harnessing their therapeutic potential reached clinical use in certain cases. In line with keeping research into cell death alive, francophone researchers from several institutions in France and Belgium established the French Cell Death Research Network (FCDRN). The research conducted by FCDRN is at the leading edge of emerging topics such as non-apoptotic functions of apoptotic effectors, paracrine effects of cell death, novel canonical and non-canonical mechanisms to induce apoptosis in cell death-resistant cancer cells or regulated forms of necrosis and the associated immunogenic response. Collectively, these various lines of research all emerged from the study of apoptosis and in the next few years will increase the mechanistic knowledge into regulated cell death and how to harness it for therapy.
- Published
- 2022
- Full Text
- View/download PDF
13. Clinical and biological features of B-cell neoplasms with CDK6 translocations: an association with a subgroup of splenic marginal zone lymphomas displaying frequent CD5 expression, prolymphocytic cells, and TP53 abnormalities.
- Author
-
Gailllard B, Cornillet-Lefebvre P, Le QH, Maloum K, Pannetier M, Lecoq-Lafon C, Grange B, Jondreville L, Michaux L, Nadal N, Ittel A, Luquet I, Struski S, Lefebvre C, Gaillard JB, Lafage-Pochitaloff M, Balducci E, Penther D, Barin C, Collonge-Rame MA, Jimenez-Poquet M, Richebourg S, Lemaire P, Defasque S, Radford-Weiss I, Bidet A, Susin SA, Nguyen-Khac F, and Chapiro E
- Subjects
- Adult, Aged, Aged, 80 and over, Bronchial Neoplasms diagnosis, Bronchial Neoplasms metabolism, Cell Differentiation, Chromosome Aberrations, Female, Genes, p53 genetics, Humans, Immunoglobulin Heavy Chains metabolism, In Situ Hybridization, Fluorescence methods, Leukemia, Lymphocytic, Chronic, B-Cell diagnosis, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy, Lymphoma, B-Cell, Marginal Zone diagnosis, Lymphoma, B-Cell, Marginal Zone drug therapy, Male, Middle Aged, Mutation, Phenotype, Survival Analysis, Tertiary Lymphoid Structures pathology, Translocation, Genetic genetics, Trisomy genetics, CD5 Antigens metabolism, Cyclin-Dependent Kinase 6 metabolism, Leukemia, Lymphocytic, Chronic, B-Cell metabolism, Lymphoma, B-Cell, Marginal Zone metabolism, Splenic Neoplasms pathology, Tumor Suppressor Protein p53 metabolism
- Abstract
A translocation involving the cyclin-dependent kinase 6 (CDK6) gene [t(CDK6)] is a rare but recurrent abnormality in B-cell neoplasms. To further characterise this aberration, we studied 57 cases; the largest series reported to date. Fluorescence in situ hybridisation analysis confirmed the involvement of CDK6 in all cases, including t(2;7)(p11;q21) immunoglobulin kappa locus (IGK)/CDK6 (n = 51), t(7;14)(q21;q32) CDK6/immunoglobulin heavy locus (IGH) (n = 2) and the previously undescribed t(7;14)(q21;q11) CDK6/T-cell receptor alpha locus (TRA)/T-cell receptor delta locus (TRD) (n = 4). In total, 10 patients were diagnosed with chronic lymphocytic leukaemia, monoclonal B-cell lymphocytosis or small lymphocytic lymphoma, and 47 had small B-cell lymphoma (SmBL) including 36 cases of marginal zone lymphoma (MZL; 34 splenic MZLs, one nodal MZL and one bronchus-associated lymphoid tissue lymphoma). In all, 18 of the 26 cytologically reviewed cases of MZL (69%) had an atypical aspect with prolymphocytic cells. Among the 47 patients with MZL/SmBL, CD5 expression was found in 26 (55%) and the tumour protein p53 (TP53) deletion in 22 (47%). The TP53 gene was mutated in 10/30 (33%); the 7q deletion was detected in only one case, and no Notch receptor 2 (NOTCH2) mutations were found. Immunoglobulin heavy-chain variable-region (IGHV) locus sequencing revealed that none harboured an IGHV1-02*04 gene. Overall survival was 82% at 10 years and not influenced by TP53 aberration. Our present findings suggest that most t(CDK6)+ neoplasms correspond to a particular subgroup of indolent marginal zone B-cell lymphomas with distinctive features., (© 2020 British Society for Haematology and John Wiley & Sons Ltd.)
- Published
- 2021
- Full Text
- View/download PDF
14. Activation of Interferon Signaling in Chronic Lymphocytic Leukemia Cells Contributes to Apoptosis Resistance via a JAK-Src/STAT3/Mcl-1 Signaling Pathway.
- Author
-
Bauvois B, Pramil E, Jondreville L, Quiney C, Nguyen-Khac F, and Susin SA
- Abstract
Besides their antiviral and immunomodulatory functions, type I (α/β) and II (γ) interferons (IFNs) exhibit either beneficial or detrimental effects on tumor progression. Chronic lymphocytic leukemia (CLL) is characterized by the accumulation of abnormal CD5
+ B lymphocytes that escape death. Drug resistance and disease relapse still occur in CLL. The triggering of IFN receptors is believed to be involved in the survival of CLL cells, but the underlying molecular mechanisms are not yet characterized. We show here that both type I and II IFNs promote the survival of primary CLL cells by counteracting the mitochondrial (intrinsic) apoptosis pathway. The survival process was associated with the upregulation of signal transducer and activator of transcription-3 (STAT3) and its target anti-apoptotic Mcl-1. Furthermore, the blockade of the STAT3/Mcl-1 pathway by pharmacological inhibitors against STAT3, TYK2 (for type I IFN) or JAK2 (for type II IFN) markedly reduced IFN-mediated CLL cell survival. Similarly, the selective Src family kinase inhibitor PP2 notably blocked IFN-mediated CLL cell survival by downregulating the protein levels of STAT3 and Mcl-1. Our work reveals a novel mechanism of resistance to apoptosis promoted by IFNs in CLL cells, whereby JAKs (TYK2, JAK2) and Src kinases activate in concert a STAT3/Mcl-1 signaling pathway. In view of current clinical developments of potent STAT3 and Mcl-1 inhibitors, a combination of conventional treatments with these inhibitors might thus constitute a new therapeutic strategy in CLL.- Published
- 2021
- Full Text
- View/download PDF
15. Mitochondrial AIF loss causes metabolic reprogramming, caspase-independent cell death blockade, embryonic lethality, and perinatal hydrocephalus.
- Author
-
Delavallée L, Mathiah N, Cabon L, Mazeraud A, Brunelle-Navas MN, Lerner LK, Tannoury M, Prola A, Moreno-Loshuertos R, Baritaud M, Vela L, Garbin K, Garnier D, Lemaire C, Langa-Vives F, Cohen-Salmon M, Fernández-Silva P, Chrétien F, Migeotte I, and Susin SA
- Subjects
- Animals, Apoptosis physiology, Caspases metabolism, Cell Respiration, Female, Fibroblasts metabolism, Genetic Engineering methods, Glycolysis genetics, Hydrocephalus metabolism, Male, Membrane Potential, Mitochondrial genetics, Membrane Potential, Mitochondrial physiology, Mice, Mice, Inbred C57BL, Mice, Inbred Strains genetics, Mitochondria metabolism, Models, Animal, Oxidative Phosphorylation, Reactive Oxygen Species metabolism, Apoptosis Inducing Factor genetics, Apoptosis Inducing Factor metabolism
- Abstract
Objectives: Apoptosis-Inducing Factor (AIF) is a protein involved in mitochondrial electron transport chain assembly/stability and programmed cell death. The relevant role of this protein is underlined because mutations altering mitochondrial AIF properties result in acute pediatric mitochondriopathies and tumor metastasis. By generating an original AIF-deficient mouse strain, this study attempted to analyze, in a single paradigm, the cellular and developmental metabolic consequences of AIF loss and the subsequent oxidative phosphorylation (OXPHOS) dysfunction., Methods: We developed a novel AIF-deficient mouse strain and assessed, using molecular and cell biology approaches, the cellular, embryonic, and adult mice phenotypic alterations. Additionally, we conducted ex vivo assays with primary and immortalized AIF knockout mouse embryonic fibroblasts (MEFs) to establish the cell death characteristics and the metabolic adaptive responses provoked by the mitochondrial electron transport chain (ETC) breakdown., Results: AIF deficiency destabilized mitochondrial ETC and provoked supercomplex disorganization, mitochondrial transmembrane potential loss, and high generation of mitochondrial reactive oxygen species (ROS). AIF
-/Y MEFs counterbalanced these OXPHOS alterations by mitochondrial network reorganization and a metabolic reprogramming toward anaerobic glycolysis illustrated by the AMPK phosphorylation at Thr172, the overexpression of the glucose transporter GLUT-4, the subsequent enhancement of glucose uptake, and the anaerobic lactate generation. A late phenotype was characterized by the activation of P53/P21-mediated senescence. Notably, approximately 2% of AIF-/Y MEFs diminished both mitochondrial mass and ROS levels and spontaneously proliferated. These cycling AIF-/Y MEFs were resistant to caspase-independent cell death inducers. The AIF-deficient mouse strain was embryonic lethal between E11.5 and E13.5 with energy loss, proliferation arrest, and increased apoptotic levels. Contrary to AIF-/Y MEFs, the AIF KO embryos were unable to reprogram their metabolism toward anaerobic glycolysis. Heterozygous AIF+/- females displayed progressive bone marrow, thymus, and spleen cellular loss. In addition, approximately 10% of AIF+/- females developed perinatal hydrocephaly characterized by brain development impairment, meningeal fibrosis, and medullar hemorrhages; those mice died 5 weeks after birth. AIF+/- with hydrocephaly exhibited loss of ciliated epithelium in the ependymal layer. This phenotype was triggered by the ROS excess. Accordingly, it was possible to diminish the occurrence of hydrocephalus AIF+/- females by supplying dams and newborns with an antioxidant in drinking water., Conclusions: In a single knockout model and at 3 different levels (cell, embryo, and adult mice) we demonstrated that by controlling the mitochondrial OXPHOS/metabolism, AIF is a key factor regulating cell differentiation and fate. Additionally, by providing new insights into the pathological consequences of mitochondrial OXPHOS dysfunction, our new findings pave the way for novel pharmacological strategies., (Copyright © 2020 The Author(s). Published by Elsevier GmbH.. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
16. Relation of Neutrophil Gelatinase-Associated Lipocalin Overexpression to the Resistance to Apoptosis of Tumor B Cells in Chronic Lymphocytic Leukemia.
- Author
-
Bauvois B, Pramil E, Jondreville L, Chapiro E, Quiney C, Maloum K, Susin SA, and Nguyen-Khac F
- Abstract
The resistance to apoptosis of chronic lymphocytic leukemia (CLL) cells partly results from the deregulated production of survival signals from leukemic cells. Despite the development of new therapies in CLL, drug resistance and disease relapse still occur. Recently, neutrophil gelatinase-associated lipocalin (NGAL), a secreted glycoprotein, has been suggested to have a critical role in the biology of tumors. Thus, we investigated the relevance of NGAL in CLL pathogenesis, analyzed the expression of its cellular receptor (NGAL-R) on malignant B cells and tested whether CLL cells are resistant to apoptosis through an autocrine process involving NGAL and NGAL-R. We observed that NGAL concentrations were elevated in the serum of CLL patients at diagnosis. After treatment (and regardless of the therapeutic regimen), serum NGAL levels normalized in CLL patients in remission but not in relapsed patients. In parallel, NGAL and NGAL-R were upregulated in leukemic cells from untreated CLL patients when compared to normal peripheral blood mononuclear cells (PBMCs), and returned to basal levels in PBMCs from patients in remission. Cultured CLL cells released endogenous NGAL. Anti-NGAL-R antibodies enhanced NGAL-R
+ leukemia cell death. Conversely, recombinant NGAL protected NGAL-R+ CLL cells against apoptosis by activating a STAT3/Mcl-1 signaling pathway. Our results suggest that NGAL and NGAL-R, overexpressed in untreated CLL, participate in the deregulation of the apoptotic machinery in CLL cells, and may be potential therapeutic clues for CLL treatment.- Published
- 2020
- Full Text
- View/download PDF
17. Genetic characterization of B-cell prolymphocytic leukemia: a prognostic model involving MYC and TP53.
- Author
-
Chapiro E, Pramil E, Diop M, Roos-Weil D, Dillard C, Gabillaud C, Maloum K, Settegrana C, Baseggio L, Lesesve JF, Yon M, Jondreville L, Lesty C, Davi F, Le Garff-Tavernier M, Droin N, Dessen P, Algrin C, Leblond V, Gabarre J, Bouzy S, Eclache V, Gaillard B, Callet-Bauchu E, Muller M, Lefebvre C, Nadal N, Ittel A, Struski S, Collonge-Rame MA, Quilichini B, Fert-Ferrer S, Auger N, Radford-Weiss I, Wagner L, Scheinost S, Zenz T, Susin SA, Bernard OA, and Nguyen-Khac F
- Subjects
- Aged, Aged, 80 and over, Chromosome Aberrations, Cytogenetic Analysis, Female, Humans, Male, Middle Aged, Prognosis, Leukemia, Prolymphocytic, B-Cell genetics, Proto-Oncogene Proteins c-myc genetics, Tumor Suppressor Protein p53 genetics
- Abstract
B-cell prolymphocytic leukemia (B-PLL) is a rare hematological disorder whose underlying oncogenic mechanisms are poorly understood. Our cytogenetic and molecular assessments of 34 patients with B-PLL revealed several disease-specific features and potential therapeutic targets. The karyotype was complex (≥3 abnormalities) in 73% of the patients and highly complex (≥5 abnormalities) in 45%. The most frequent chromosomal aberrations were translocations involving MYC [t(MYC)] (62%), deletion (del)17p (38%), trisomy (tri)18 (30%), del13q (29%), tri3 (24%), tri12 (24%), and del8p (23%). Twenty-six (76%) of the 34 patients exhibited an MYC aberration, resulting from mutually exclusive translocations or gains. Whole-exome sequencing revealed frequent mutations in TP53, MYD88, BCOR, MYC, SF3B1, SETD2, CHD2, CXCR4, and BCLAF1. The majority of B-PLL used the IGHV3 or IGHV4 subgroups (89%) and displayed significantly mutated IGHV genes (79%). We identified 3 distinct cytogenetic risk groups: low risk (no MYC aberration), intermediate risk (MYC aberration but no del17p), and high risk (MYC aberration and del17p) (P = .0006). In vitro drug response profiling revealed that the combination of a B-cell receptor or BCL2 inhibitor with OTX015 (a bromodomain and extra-terminal motif inhibitor targeting MYC) was associated with significantly lower viability of B-PLL cells harboring a t(MYC). We concluded that cytogenetic analysis is a useful diagnostic and prognostic tool in B-PLL. Targeting MYC may be a useful treatment option in this disease., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
18. Targeting chronic lymphocytic leukemia with N-methylated thrombospondin-1-derived peptides overcomes drug resistance.
- Author
-
Pramil E, Herbi Bastian L, Denèfle T, Nemati F, Xiao M, Lardé E, Maloum K, Roos-Weil D, Chapiro E, Le Garff-Tavernier M, Davi F, Decaudin D, Sarfati M, Nguyen-Khac F, Merle-Béral H, Karoyan P, and Susin SA
- Subjects
- Animals, Apoptosis drug effects, Caspases metabolism, Cell Cycle Checkpoints drug effects, Cell Death drug effects, Cell Line, Tumor, Cyclic AMP metabolism, Cyclic AMP-Dependent Protein Kinases metabolism, Disease Models, Animal, Female, Humans, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy, Leukemia, Lymphocytic, Chronic, B-Cell metabolism, Leukemia, Lymphocytic, Chronic, B-Cell pathology, Mice, Mitochondria drug effects, Mitochondria metabolism, Models, Molecular, Molecular Mimicry, Peptides chemistry, Signal Transduction drug effects, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacology, Drug Resistance, Neoplasm drug effects, Peptides pharmacology, Thrombospondin 1 chemistry
- Abstract
Chronic lymphocytic leukemia (CLL), the most common adulthood leukemia in Western countries, is a very heterogeneous disease characterized by a peripheral accumulation of abnormal CD5+ B lymphocytes in the immune system. Despite new therapeutic developments, there remains an unmet medical need for CLL. Here, we demonstrate that the use of N-methylated thrombospondin-1 (TSP-1)-derived peptides is an efficient way to kill the malignant CLL cells, including those from high-risk individuals with poor clinical prognosis, del11q, del17p, 2p gain, or complex karyotype. PKT16, our hit N-methylated peptide, triggers the elimination of the leukemic cells, sparing the nontumor cells, including the hematopoietic precursors, and reduces the in vivo tumor burden of a CLL-xenograft mice model. A complementary analysis underscores the improved cytotoxic efficiency of PKT16 compared with the previously described TSP-1-derived probes, such as PKHB1. PKT16 elicits an original caspase-independent programmed necrotic mode of cell death, different from necroptosis or ferroptosis, implicating an intracellular Ca2+ deregulation that provokes mitochondrial damage, cell cycle arrest, and the specific death of the malignant CLL cells. The activation of the Gαi proteins and the subsequent drop of cyclic adenosine monophosphate levels and protein kinase A activity regulate this cytotoxic cascade. Remarkably, PKT16 induces the molecular hallmarks of immunogenic cell death, as defined by the calreticulin plasma membrane exposure and the release of adenosine triphosphate and high-mobility group box 1 protein from the dying CLL cells. Thus, PKT16 appears to be able to stimulate an anticancer in vivo immune response. Collectively, our results pave the way toward the development of an efficient strategy against CLL., (© 2019 by The American Society of Hematology.)
- Published
- 2019
- Full Text
- View/download PDF
19. Homotrimerization Approach in the Design of Thrombospondin-1 Mimetic Peptides with Improved Potency in Triggering Regulated Cell Death of Cancer Cells.
- Author
-
Denèfle T, Pramil E, Gómez-Morales L, Levasseur MD, Lardé E, Newton C, Herry K, Herbi L, Lamotte Y, Odile E, Ancellin N, Grondin P, Martinez-Torres AC, Viviani F, Merle-Beral H, Lequin O, Susin SA, and Karoyan P
- Subjects
- A549 Cells, Cell Death drug effects, Cell Line, Tumor, Cell Survival drug effects, Dose-Response Relationship, Drug, Humans, Peptides chemical synthesis, Protein Conformation, Protein Stability, Structure-Activity Relationship, Thrombospondin 1 pharmacology, Drug Design, Peptides chemistry, Peptides pharmacology, Thrombospondin 1 chemistry
- Abstract
In order to optimize the potency of the first serum-stable peptide agonist of CD47 (PKHB1) in triggering regulated cell death of cancer cells, we designed a maturation process aimed to mimic the trimeric structure of the thrombospondin-1/CD47 binding epitope. For that purpose, an N-methylation scan of the PKHB1 sequence was realized to prevent peptide aggregation. Structural and pharmacological analyses were conducted in order to assess the conformational impact of these chemical modifications on the backbone structure and the biological activity. This structure-activity relationship study led to the discovery of a highly soluble N-methylated peptide that we termed PKT16. Afterward, this monomer was used for the design of a homotrimeric peptide mimic that we termed [PKT16]
3 , which proved to be 10-fold more potent than its monomeric counterpart. A pharmacological evaluation of [PKT16]3 in inducing cell death of adherent (A549) and nonadherent (MEC-1) cancer cell lines was also performed.- Published
- 2019
- Full Text
- View/download PDF
20. Gain of the short arm of chromosome 2 (2p gain) has a significant role in drug-resistant chronic lymphocytic leukemia.
- Author
-
Kostopoulou F, Gabillaud C, Chapiro E, Grange B, Tran J, Bouzy S, Degaud M, Ghamlouch H, Le Garff-Tavernier M, Maloum K, Choquet S, Leblond V, Gabarre J, Lavaud A, Morel V, Roos-Weil D, Uzunov M, Guieze R, Bernard OA, Susin SA, Tournilhac O, and Nguyen-Khac F
- Subjects
- Adult, Aged, Antineoplastic Agents therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Chromosome Aberrations, Female, Humans, Immunoglobulin Variable Region genetics, Immunophenotyping, In Situ Hybridization, Fluorescence, Karyotyping, Leukemia, Lymphocytic, Chronic, B-Cell diagnosis, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy, Leukemia, Lymphocytic, Chronic, B-Cell mortality, Longitudinal Studies, Male, Middle Aged, Mutation, Polymorphism, Single Nucleotide, Prognosis, Protein Kinase Inhibitors pharmacology, Protein Kinase Inhibitors therapeutic use, Recurrence, Time-to-Treatment, Treatment Outcome, Antineoplastic Agents pharmacology, Chromosome Duplication, Chromosomes, Human, Pair 2, Drug Resistance, Neoplasm genetics, Leukemia, Lymphocytic, Chronic, B-Cell genetics
- Abstract
The different types of drug resistance encountered in chronic lymphocytic leukemia (CLL) cannot be fully accounted for by the 17p deletion (and/or TP53 mutation), a complex karyotype (CK), immunoglobulin heavy-chain variable region genes (IGHV) status and gene mutations. Hence, we sought to assess the associations between recurrent genomic abnormalities in CLL and the disease's development and outcome. To this end, we analyzed 64 samples from patients with CLL and gain of the short arm of chromosome 2 (2p+), which is frequent in late-stage and relapsed/refractory CLL. We found that fludarabine/cyclophosphamide/rituximab (a common first-line treatment in CLL) is not effective in removing the 2p+ clone - even in samples lacking a CK, the 17p deletion or unmutated IGHV. Our results suggest strongly that patients with CLL should be screened for 2p+ (using karyotyping and fluorescence in situ hybridization) before a treatment option is chosen. Longer follow-up is now required to evaluate bendamustine-rituximab, ibrutinib, and idelalisib-rituximab treatments., (© 2019 The Authors. Cancer Medicine published by John Wiley & Sons Ltd.)
- Published
- 2019
- Full Text
- View/download PDF
21. CD47 agonist peptide PKHB1 induces immunogenic cell death in T-cell acute lymphoblastic leukemia cells.
- Author
-
Uscanga-Palomeque AC, Calvillo-Rodríguez KM, Gómez-Morales L, Lardé E, Denèfle T, Caballero-Hernández D, Merle-Béral H, Susin SA, Karoyan P, Martínez-Torres AC, and Rodríguez-Padilla C
- Subjects
- Animals, CD47 Antigen metabolism, Calcium metabolism, Cell Death drug effects, Cell Line, Tumor, Female, Humans, Kaplan-Meier Estimate, Leukemia, Experimental drug therapy, Leukemia, Experimental metabolism, Leukemia, Experimental pathology, Mice, Inbred BALB C, Peptides chemistry, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma metabolism, Precursor T-Cell Lymphoblastic Leukemia-Lymphoma pathology, Thrombospondin 1 chemistry, Apoptosis drug effects, CD47 Antigen agonists, Membrane Potential, Mitochondrial drug effects, Peptides pharmacology
- Abstract
T-cell acute lymphoblastic leukemia (T-ALL) has a poor prognosis derived from its genetic heterogeneity, which translates to a high chemoresistance. Recently, our workgroup designed thrombospondin-1-derived CD47 agonist peptides and demonstrated their ability to induce cell death in chronic lymphocytic leukemia. Encouraged by these promising results, we evaluated cell death induced by PKHB1 (the first-described serum-stable CD47-agonist peptide) on CEM and MOLT-4 human cell lines (T-ALL) and on one T-murine tumor lymphoblast cell-line (L5178Y-R), also assessing caspase and calcium dependency and mitochondrial membrane potential. Additionally, we evaluated selectivity for cancer cell lines by analyzing cell death and viability of human and murine non-tumor cells after CD47 activation. In vivo, we determined that PKHB1-treatment in mice bearing the L5178Y-R cell line increased leukocyte cell count in peripheral blood and lymphoid organs while recruiting leukocytes to the tumor site. To analyze whether CD47 activation induced immunogenic cell death (ICD), we evaluated damage-associated molecular patterns (DAMP) exposure (calreticulin, CRT) and release (ATP, heat shock proteins 70 and 90, high-mobility group box 1, CRT). Furthermore, we gave prophylactic antitumor vaccination, determining immunological memory. Our data indicate that PKHB1 induces caspase-independent and calcium-dependent cell death in leukemic cells while sparing non-tumor murine and human cells. Moreover, our results show that PKHB1 can induce ICD in leukemic cells as it induces CRT exposure and DAMP release in vitro, and prophylactic vaccinations inhibit tumor establishment in vivo. Together, our results improve the knowledge of CD47 agonist peptides potential as therapeutic tools to treat leukemia., (© 2018 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association.)
- Published
- 2019
- Full Text
- View/download PDF
22. Revisiting Neutrophil Gelatinase-Associated Lipocalin (NGAL) in Cancer: Saint or Sinner?
- Author
-
Bauvois B and Susin SA
- Abstract
Human neutrophil gelatinase-associated lipocalin (NGAL) is a glycoprotein present in a wide variety of tissues and cell types. NGAL exists as a 25 kDa monomer, a 46 kDa homodimer (the most abundant form in healthy subjects) and a 130 kDa disulfide-linked heterodimer bound to latent matrix metalloproteinase-9. Dysregulated expression of NGAL in human malignancies suggests its value as a clinical marker. A growing body of evidence is highlighting NGAL's paradoxical (i.e., both beneficial and detrimental) effects on cellular processes associated with tumor development (proliferation, survival, migration, invasion, and multidrug resistance). At least two distinct cell surface receptors are identified for NGAL. This review (i) summarizes our current knowledge of NGAL's expression profiles in solid tumors and leukemias, and (ii) critically evaluates the beneficial and detrimental activities of NGAL having been documented in a diverse range of cancer-derived cell lines. A better understanding of the causal relationships between NGAL dysregulation and tumor development will require a fine analysis of the molecular aspects and biological role(s) of NGAL both in primary tumors and at different stages of disease. Having an accurate picture of NGAL's contribution to tumor progression is a prerequisite for attempting to modulate this protein as a putative therapeutic target.
- Published
- 2018
- Full Text
- View/download PDF
23. Mitochondrial OXPHOS influences immune cell fate: lessons from hematopoietic AIF-deficient and NDUFS4-deficient mouse models.
- Author
-
Bertaux A, Cabon L, Brunelle-Navas MN, Bouchet S, Nemazanyy I, and Susin SA
- Subjects
- Animals, Apoptosis Inducing Factor metabolism, Bone Marrow Cells metabolism, Electron Transport Complex I metabolism, Gene Deletion, Hematopoietic Stem Cells metabolism, Mice, Models, Animal, Thymocytes metabolism, Apoptosis Inducing Factor deficiency, Electron Transport Complex I deficiency, Hematopoietic Stem Cells immunology, Hematopoietic Stem Cells pathology, Mitochondria metabolism, Oxidative Phosphorylation
- Published
- 2018
- Full Text
- View/download PDF
24. AIF loss deregulates hematopoiesis and reveals different adaptive metabolic responses in bone marrow cells and thymocytes.
- Author
-
Cabon L, Bertaux A, Brunelle-Navas MN, Nemazanyy I, Scourzic L, Delavallée L, Vela L, Baritaud M, Bouchet S, Lopez C, Quang Van V, Garbin K, Chateau D, Gilard F, Sarfati M, Mercher T, Bernard OA, and Susin SA
- Subjects
- Animals, B-Lymphocytes cytology, Hematopoietic Stem Cells cytology, Mice, Mice, Knockout, Mitochondria genetics, Mitochondria metabolism, Reactive Oxygen Species metabolism, Thymocytes cytology, Apoptosis Inducing Factor deficiency, B-Lymphocytes metabolism, Hematopoiesis, Hematopoietic Stem Cells metabolism, Oxidative Phosphorylation, Thymocytes metabolism
- Abstract
Mitochondrial metabolism is a tightly regulated process that plays a central role throughout the lifespan of hematopoietic cells. Herein, we analyze the consequences of the mitochondrial oxidative phosphorylation (OXPHOS)/metabolism disorder associated with the cell-specific hematopoietic ablation of apoptosis-inducing factor (AIF). AIF-null (AIF
- /Y ) mice developed pancytopenia that was associated with hypocellular bone marrow (BM) and thymus atrophy. Although myeloid cells were relatively spared, the B-cell and erythroid lineages were altered with increased frequencies of precursor B cells, pro-erythroblasts I, and basophilic erythroblasts II. T-cell populations were dramatically reduced with a thymopoiesis blockade at a double negative (DN) immature state, with DN1 accumulation and delayed DN2/DN3 and DN3/DN4 transitions. In BM cells, the OXPHOS/metabolism dysfunction provoked by the loss of AIF was counterbalanced by the augmentation of the mitochondrial biogenesis and a shift towards anaerobic glycolysis. Nevertheless, in a caspase-independent process, the resulting excess of reactive oxygen species compromised the viability of the hematopoietic stem cells (HSC) and progenitors. This led to the progressive exhaustion of the HSC pool, a reduced capacity of the BM progenitors to differentiate into colonies in methylcellulose assays, and the absence of cell-autonomous HSC repopulating potential in vivo. In contrast to BM cells, AIF- /Y thymocytes compensated for the OXPHOS breakdown by enhancing fatty acid β-oxidation. By over-expressing CPT1, ACADL and PDK4, three key enzymes facilitating fatty acid β-oxidation (e.g., palmitic acid assimilation), the AIF- /Y thymocytes retrieved the ATP levels of the AIF+/Y cells. As a consequence, it was possible to significantly reestablish AIF- /Y thymopoiesis in vivo by feeding the animals with a high-fat diet complemented with an antioxidant. Overall, our data reveal that the mitochondrial signals regulated by AIF are critical to hematopoietic decision-making. Emerging as a link between mitochondrial metabolism and hematopoietic cell fate, AIF-mediated OXPHOS regulation represents a target for the development of new immunomodulatory therapeutics.- Published
- 2018
- Full Text
- View/download PDF
25. "Double-hit" chronic lymphocytic leukemia: An aggressive subgroup with 17p deletion and 8q24 gain.
- Author
-
Chapiro E, Lesty C, Gabillaud C, Durot E, Bouzy S, Armand M, Le Garff-Tavernier M, Bougacha N, Struski S, Bidet A, Laharanne E, Barin C, Veronese L, Prié N, Eclache V, Gaillard B, Michaux L, Lefebvre C, Gaillard JB, Terré C, Penther D, Bastard C, Nadal N, Fert-Ferrer S, Auger N, Godon C, Sutton L, Tournilhac O, Susin SA, and Nguyen-Khac F
- Subjects
- Abnormal Karyotype, Adult, Aged, Aged, 80 and over, Disease-Free Survival, Female, Genes, p53, Humans, In Situ Hybridization, Fluorescence, Kaplan-Meier Estimate, Leukemia, Lymphocytic, Chronic, B-Cell mortality, Leukemia, Lymphocytic, Chronic, B-Cell pathology, Male, Middle Aged, Neoplasm Invasiveness genetics, Prognosis, Retrospective Studies, Chromosome Deletion, Chromosomes, Human, Pair 17 ultrastructure, Chromosomes, Human, Pair 8 ultrastructure, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Translocation, Genetic, Trisomy
- Abstract
Chronic lymphocytic leukemia (CLL) with 17p deletion (17p-) is associated with a lack of response to standard treatment and thus the worst possible clinical outcome. Various chromosomal abnormalities (including unbalanced translocations, deletions, ring chromosomes and isochromosomes) result in the loss of 17p and one copy of the TP53 gene. The objective of the present study was to determine whether the type of chromosomal abnormality leading to 17p- and the additional aberrations influenced the prognosis in a series of 195 patients with 17p-CLL. Loss of 17p resulted primarily from an unbalanced translocation (70%) with several chromosome partners (the most frequent being chromosome 18q), followed by deletion 17p (23%), monosomy 17 (8%), isochromosome 17q [i(17q)] (5%) and a ring chromosome 17 (2%). In a univariate analysis, monosomy 17, a highly complex karyotype (≥5 abnormalities), and 8q24 gain were associated with poor treatment-free survival, and i(17q) (P = .04), unbalanced translocations (P = .03) and 8q24 gain (P = .001) were significantly associated with poor overall survival. In a multivariate analysis, 8q24 gain remained a significant predictor of poor overall survival. We conclude that 17p deletion and 8q24 gain have a synergistic impact on outcome, and so patients with this "double-hit" CLL have a particularly poor prognosis. Systematic, targeting screening for 8q24 gain should therefore be considered in cases of 17p- CLL., (© 2017 Wiley Periodicals, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
26. Gain in the short arm of chromosome 2 (2p+) induces gene overexpression and drug resistance in chronic lymphocytic leukemia: analysis of the central role of XPO1.
- Author
-
Cosson A, Chapiro E, Bougacha N, Lambert J, Herbi L, Cung HA, Algrin C, Keren B, Damm F, Gabillaud C, Brunelle-Navas MN, Davi F, Merle-Béral H, Le Garff-Tavernier M, Roos-Weil D, Choquet S, Uzunov M, Morel V, Leblond V, Maloum K, Lepretre S, Feugier P, Lesty C, Lejeune J, Sutton L, Landesman Y, Susin SA, and Nguyen-Khac F
- Subjects
- Apoptosis, Drug Resistance, Neoplasm drug effects, Humans, Hydrazines pharmacology, Leukemia, Lymphocytic, Chronic, B-Cell genetics, Triazoles pharmacology, Exportin 1 Protein, Chromosomes, Human, Pair 2, Drug Resistance, Neoplasm genetics, Gene Expression Regulation, Leukemic, Hydrazines therapeutic use, Karyopherins genetics, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy, Receptors, Cytoplasmic and Nuclear genetics, Triazoles therapeutic use
- Published
- 2017
- Full Text
- View/download PDF
27. Thrombospondin-1 Mimetic Agonist Peptides Induce Selective Death in Tumor Cells: Design, Synthesis, and Structure-Activity Relationship Studies.
- Author
-
Denèfle T, Boullet H, Herbi L, Newton C, Martinez-Torres AC, Guez A, Pramil E, Quiney C, Pourcelot M, Levasseur MD, Lardé E, Moumné R, Ogi FX, Grondin P, Merle-Beral H, Lequin O, Susin SA, and Karoyan P
- Subjects
- Amino Acid Sequence, Apoptosis drug effects, Cell Line, Tumor, Humans, Models, Molecular, Neoplasms drug therapy, Neoplasms metabolism, Structure-Activity Relationship, Thrombospondin 1 metabolism, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacology, Peptides chemistry, Peptides pharmacology, Thrombospondin 1 agonists
- Abstract
Thrombospondin-1 (TSP-1) is a glycoprotein considered as a key actor within the tumor microenvironment. Its binding to CD47, a cell surface receptor, triggers programmed cell death. Previous studies allowed the identification of 4N1K decapeptide derived from the TSP-1/CD47 binding epitope. Here, we demonstrate that this peptide is able to induce selective apoptosis of various cancer cell lines while sparing normal cells. A structure-activity relationship study led to the design of the first serum stable TSP-1 mimetic agonist peptide able to trigger selective programmed cell death (PCD) of at least lung, breast, and colorectal cancer cells. Altogether, these results will be of valuable interest for further investigation in the design of potent CD47 agonist peptides, opening new perspectives for the development of original anticancer therapies.
- Published
- 2016
- Full Text
- View/download PDF
28. Key Residues Regulating the Reductase Activity of the Human Mitochondrial Apoptosis Inducing Factor.
- Author
-
Villanueva R, Ferreira P, Marcuello C, Usón A, Miramar MD, Peleato ML, Lostao A, Susin SA, and Medina M
- Subjects
- Amino Acid Sequence, Apoptosis Inducing Factor genetics, Catalytic Domain, Conserved Sequence, Humans, Models, Molecular, Molecular Sequence Data, Mutagenesis, Site-Directed, Mutation, NAD metabolism, Protein Multimerization, Protein Structure, Quaternary, Apoptosis Inducing Factor chemistry, Apoptosis Inducing Factor metabolism, Mitochondria enzymology
- Abstract
The human Apoptosis Inducing Factor (hAIF) is a bifunctional NAD(P)H-dependent flavoreductase involved in both mitochondrial energy metabolism and caspase-independent cell death. Even though several studies indicate that both functions are redox controlled by NADH binding, the exact role of hAIF as a reductase in healthy mitochondria remains unknown. Upon reduction by NADH, hAIF dimerizes and produces very stable flavin/nicotinamide charge transfer complexes (CTC), by stacking of the oxidized nicotinamide moiety of the NAD(+) coenzyme against the re-face of the reduced flavin ring of its FAD cofactor. Such complexes are critical to restrict the hAIF efficiency as a reductase. The molecular basis of the hAIF reductase activity is here investigated by analyzing the role played by residues contributing to the interaction of the FAD isoalloxazine ring and of the nicotinamide moiety of NADH at the active site. Mutations at K177 and E314 produced drastic effects on the hAIF ability to retain the FAD cofactor, indicating that these residues are important to set up the holo-enzyme active site conformation. Characterization of P173G hAIF indicates that the stacking of P173 against the isoalloxazine ring is relevant to determine the flavin environment and to modulate the enzyme affinity for NADH. Finally, the properties of the F310G and H454S hAIF mutants indicate that these two positions contribute to form a compact active site essential for NADH binding, CTC stabilization, and NAD(+) affinity for the reduced state of hAIF. These features are key determinants of the particular behavior of hAIF as a NADH-dependent oxidoreductase.
- Published
- 2015
- Full Text
- View/download PDF
29. CD47 agonist peptides induce programmed cell death in refractory chronic lymphocytic leukemia B cells via PLCγ1 activation: evidence from mice and humans.
- Author
-
Martinez-Torres AC, Quiney C, Attout T, Boullet H, Herbi L, Vela L, Barbier S, Chateau D, Chapiro E, Nguyen-Khac F, Davi F, Le Garff-Tavernier M, Moumné R, Sarfati M, Karoyan P, Merle-Béral H, Launay P, and Susin SA
- Subjects
- Aged, Aged, 80 and over, Animals, Female, Humans, Leukemia, Lymphocytic, Chronic, B-Cell blood, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy, Male, Mice, Mice, Inbred NOD, Middle Aged, Peptides therapeutic use, Thrombospondin 1 therapeutic use, Apoptosis drug effects, B-Lymphocytes metabolism, CD47 Antigen metabolism, Drug Resistance, Neoplasm, Leukemia, Lymphocytic, Chronic, B-Cell metabolism, Peptides pharmacology, Phospholipase C gamma metabolism
- Abstract
Background: Chronic lymphocytic leukemia (CLL), the most common adulthood leukemia, is characterized by the accumulation of abnormal CD5+ B lymphocytes, which results in a progressive failure of the immune system. Despite intense research efforts, drug resistance remains a major cause of treatment failure in CLL, particularly in patients with dysfunctional TP53. The objective of our work was to identify potential approaches that might overcome CLL drug refractoriness by examining the pro-apoptotic potential of targeting the cell surface receptor CD47 with serum-stable agonist peptides., Methods and Findings: In peripheral blood samples collected from 80 patients with CLL with positive and adverse prognostic features, we performed in vitro genetic and molecular analyses that demonstrate that the targeting of CD47 with peptides derived from the C-terminal domain of thrombospondin-1 efficiently kills the malignant CLL B cells, including those from high-risk individuals with a dysfunctional TP53 gene, while sparing the normal T and B lymphocytes from the CLL patients. Further studies reveal that the differential response of normal B lymphocytes, collected from 20 healthy donors, and leukemic B cells to CD47 peptide targeting results from the sustained activation in CLL B cells of phospholipase C gamma-1 (PLCγ1), a protein that is significantly over-expressed in CLL. Once phosphorylated at tyrosine 783, PLCγ1 enables a Ca2+-mediated, caspase-independent programmed cell death (PCD) pathway that is not down-modulated by the lymphocyte microenvironment. Accordingly, down-regulation of PLCγ1 or pharmacological inhibition of PLCγ1 phosphorylation abolishes CD47-mediated killing. Additionally, in a CLL-xenograft model developed in NOD/scid gamma mice, we demonstrate that the injection of CD47 agonist peptides reduces tumor burden without inducing anemia or toxicity in blood, liver, or kidney. The limitations of our study are mainly linked to the affinity of the peptides targeting CD47, which might be improved to reach the standard requirements in drug development, and the lack of a CLL animal model that fully mimics the human disease., Conclusions: Our work provides substantial progress in (i) the development of serum-stable CD47 agonist peptides that are highly effective at inducing PCD in CLL, (ii) the understanding of the molecular events regulating a novel PCD pathway that overcomes CLL apoptotic avoidance, (iii) the identification of PLCγ1 as an over-expressed protein in CLL B cells, and (iv) the description of a novel peptide-based strategy against CLL.
- Published
- 2015
- Full Text
- View/download PDF
30. Structural insights into the coenzyme mediated monomer-dimer transition of the pro-apoptotic apoptosis inducing factor.
- Author
-
Ferreira P, Villanueva R, Martínez-Júlvez M, Herguedas B, Marcuello C, Fernandez-Silva P, Cabon L, Hermoso JA, Lostao A, Susin SA, and Medina M
- Subjects
- Apoptosis Inducing Factor genetics, Crystallography, X-Ray, Humans, Kinetics, Models, Molecular, Mutation, Promoter Regions, Genetic, Protein Conformation, Protein Multimerization, Apoptosis, Apoptosis Inducing Factor chemistry, NAD chemistry
- Abstract
The apoptosis-inducing factor (AIF) is a mitochondrial-flavoprotein that, after cell death induction, is distributed to the nucleus to mediate chromatinolysis. In mitochondria, AIF is present in a monomer-dimer equilibrium that after reduction by NADH gets displaced toward the dimer. The crystal structure of the human AIF (hAIF):NAD(H)-bound dimer revealed one FAD and, unexpectedly, two NAD(H) molecules per protomer. A 1:2 hAIF:NAD(H) binding stoichiometry was additionally confirmed in solution by using surface plasmon resonance. The here newly discovered NAD(H)-binding site includes residues mutated in human disorders, and accommodation of the coenzyme in it requires restructuring of a hAIF portion within the 509-560 apoptogenic segment. Disruption of interactions at the dimerization surface by production of the hAIF E413A/R422A/R430A mutant resulted in a nondimerizable variant considerably less efficiently stabilizing charge-transfer complexes upon coenzyme reduction than WT hAIF. These data reveal that the coenzyme-mediated monomer-dimer transition of hAIF modulates the conformation of its C-terminal proapoptotic domain, as well as its mechanism as reductase. These observations suggest that both the mitochondrial and apoptotic functions of hAIF are interconnected and coenzyme controlled: a key information in the understanding of the physiological role of AIF in the cellular life and death cycle.
- Published
- 2014
- Full Text
- View/download PDF
31. [Programmed cell death comes in many flavors].
- Author
-
Cabon L, Martinez-Torres AC, and Susin SA
- Subjects
- Animals, Anoikis physiology, Autophagy, Cell Death physiology, Entosis physiology, Humans, Necrosis, Signal Transduction, Apoptosis physiology
- Abstract
Apoptosis is nowadays what comes first to your scientist mind when someone mentions cellular suicide. However this is not the sole form of programmed cell death and many other alternative or atypical pathways have now been described. These pathways are indeed rather preferred to apoptosis in some instances based on tissue origin, cell type or development stage of the target cell. In this review, we describe many different programmed cell death subtypes according to their characteristics. Discrete, brutal, final or singular cell death pathways all participate in the elimination of unwanted, damaged or dangerous cells in organisms hence contributing to our knowledge of this particular feature of living beings: dying! Through description of anoikis, necroptosis, entosis, netosis, pyroptosis or ferroptosis, we have no choice but to realize that programmed cell death comes in many flavors., (© 2013 médecine/sciences – Inserm.)
- Published
- 2013
- Full Text
- View/download PDF
32. AIF-mediated caspase-independent necroptosis requires ATM and DNA-PK-induced histone H2AX Ser139 phosphorylation.
- Author
-
Baritaud M, Cabon L, Delavallée L, Galán-Malo P, Gilles ME, Brunelle-Navas MN, and Susin SA
- Subjects
- Animals, Apoptosis drug effects, Ataxia Telangiectasia Mutated Proteins, Cell Cycle Proteins antagonists & inhibitors, Cell Line, Chromatin metabolism, DNA Breaks, Double-Stranded drug effects, DNA-Activated Protein Kinase antagonists & inhibitors, DNA-Binding Proteins antagonists & inhibitors, GTPase-Activating Proteins genetics, GTPase-Activating Proteins metabolism, Histones genetics, Kinetics, Methylnitronitrosoguanidine pharmacology, Mice, Phosphatidylinositol 3-Kinases metabolism, Phosphorylation, Protein Serine-Threonine Kinases antagonists & inhibitors, Tumor Suppressor Proteins antagonists & inhibitors, Apoptosis Inducing Factor metabolism, Caspases metabolism, Cell Cycle Proteins metabolism, DNA-Activated Protein Kinase metabolism, DNA-Binding Proteins metabolism, Histones metabolism, Protein Serine-Threonine Kinases metabolism, Tumor Suppressor Proteins metabolism
- Abstract
The alkylating DNA-damage agent N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) induces a form of caspase-independent necroptosis implicating the mitochondrial flavoprotein apoptosis-inducing factor (AIF). Following the activation of PARP-1 (poly(ADP-ribose) polymerase-1), calpains, BID (BH3 interacting domain death agonist), and BAX (Bcl-2-associated X protein), the apoptogenic form of AIF (tAIF) is translocated to the nucleus where, associated with Ser139-phosphorylated histone H2AX (γH2AX), it creates a DNA-degrading complex that provokes chromatinolysis and cell death by necroptosis. The generation of γH2AX is crucial for this form of cell death, as mutation of H2AX Ser139 to Ala or genetic ablation of H2AX abolish both chromatinolysis and necroptosis. On the contrary, reintroduction of H2AX-wt or the phosphomimetic H2AX mutant (H2AX-S139E) into H2AX(-/-) cells resensitizes to MNNG-triggered necroptosis. Employing a pharmacological approach and gene knockout cells, we also demonstrate in this paper that the phosphatidylinositol-3-OH kinase-related kinases (PIKKs) ATM (ataxia telangiectasia mutated) and DNA-dependent protein kinase (DNA-PK) mediate γH2AX generation and, consequently, MNNG-induced necroptosis. By contrast, H2AX phosphorylation is not regulated by ATR or other H2AX-related kinases, such as JNK. Interestingly, ATM and DNA-PK phosphorylate H2AX at Ser139 in a synergistic manner with different kinetics of activation. Early after MNNG treatment, ATM generates γH2AX. Further, DNA-PK contributes to H2AX Ser139 phosphorylation. In revealing the pivotal role of PIKKs in MNNG-induced cell death, our data uncover a milestone in the mechanisms regulating AIF-mediated caspase-independent necroptosis.
- Published
- 2012
- Full Text
- View/download PDF
33. CD47(high) expression on CD4 effectors identifies functional long-lived memory T cell progenitors.
- Author
-
Van VQ, Raymond M, Baba N, Rubio M, Wakahara K, Susin SA, and Sarfati M
- Subjects
- Animals, CD4-Positive T-Lymphocytes cytology, CD4-Positive T-Lymphocytes metabolism, CD47 Antigen biosynthesis, CD47 Antigen genetics, Mice, Mice, Knockout, Precursor Cells, T-Lymphoid cytology, Precursor Cells, T-Lymphoid metabolism, CD4-Positive T-Lymphocytes immunology, CD47 Antigen immunology, Gene Expression Regulation physiology, Immunity, Cellular physiology, Immunologic Memory physiology, Precursor Cells, T-Lymphoid immunology
- Abstract
T cell memory is the hallmark of adaptive immunity. Central questions are to determine which cells among proliferating effector T cells will live beyond the crash of the immune response (IR) and develop into functional memory T cells. CD47, considered as a marker of self, is implicated in cell death, cell elimination, and in the inflammatory response. We report in this article that CD47 expression was transiently regulated on Ag-specific CD4 T cells, that is, from CD47(high) to CD47(low) to CD47(high), during the course of the in vivo IR. Specifically, CD47(high) status marked central memory CD4 T cell precursors at an early time point of the IR. By contrast, cytokine production was a functional attribute restricted to CD47(high), but not CD47(low), polyclonal effector CD4 T cells during recall responses in an experimental model of chronic airway inflammatory disease. Passive transfer of CD47(high), but not CD47(low), CD4 T cells in nonlymphopenic naive mice generated long-lived memory T cells capable of anamnestic responses. We conclude that CD47(high) status on CD4 T cells identifies functional long-lived memory T cell progenitors.
- Published
- 2012
- Full Text
- View/download PDF
34. BID regulates AIF-mediated caspase-independent necroptosis by promoting BAX activation.
- Author
-
Cabon L, Galán-Malo P, Bouharrour A, Delavallée L, Brunelle-Navas MN, Lorenzo HK, Gross A, and Susin SA
- Subjects
- Animals, Apoptosis Regulatory Proteins metabolism, Bcl-2-Like Protein 11, Calpain metabolism, Caspases metabolism, Cell Survival drug effects, DNA Damage, Embryo, Mammalian cytology, Fibroblasts cytology, Fibroblasts drug effects, Fibroblasts metabolism, Lentivirus genetics, Membrane Proteins metabolism, Methylnitronitrosoguanidine pharmacology, Mice, Models, Biological, Necrosis, Poly(ADP-ribose) Polymerases metabolism, Proto-Oncogene Proteins metabolism, Transduction, Genetic, bcl-Associated Death Protein metabolism, Apoptosis drug effects, Apoptosis Inducing Factor metabolism, BH3 Interacting Domain Death Agonist Protein metabolism, bcl-2-Associated X Protein metabolism
- Abstract
Alkylating DNA-damage agents such as N-methyl-N'-nitro-N'-nitrosoguanidine (MNNG) trigger necroptosis, a newly defined form of programmed cell death (PCD) managed by receptor interacting protein kinases. This caspase-independent mode of cell death involves the sequential activation of poly(ADP-ribose) polymerase-1 (PARP-1), calpains, BAX and AIF, which redistributes from mitochondria to the nucleus to promote chromatinolysis. We have previously demonstrated that the BAX-mediated mitochondrial release of AIF is a critical step in MNNG-mediated necroptosis. However, the mechanism regulating BAX activation in this PCD is poorly understood. Employing mouse embryonic knockout cells, we reveal that BID controls BAX activation in AIF-mediated necroptosis. Indeed, BID is a link between calpains and BAX in this mode of cell death. Therefore, even if PARP-1 and calpains are activated after MNNG treatment, BID genetic ablation abolishes both BAX activation and necroptosis. These PCD defects are reversed by reintroducing the BID-wt cDNA into the BID(-/-) cells. We also demonstrate that, after MNNG treatment, BID is directly processed into tBID by calpains. In this way, calpain non-cleavable BID proteins (BID-G70A or BID-Δ68-71) are unable to promote BAX activation and necroptosis. Once processed, tBID localizes in the mitochondria of MNNG-treated cells, where it can facilitate BAX activation and PCD. Altogether, our data reveal that, as in caspase-dependent apoptosis, BH3-only proteins are key regulators of caspase-independent necroptosis.
- Published
- 2012
- Full Text
- View/download PDF
35. AIF-mediated caspase-independent necroptosis: a new chance for targeted therapeutics.
- Author
-
Delavallée L, Cabon L, Galán-Malo P, Lorenzo HK, and Susin SA
- Subjects
- Amino Acid Sequence, Animals, Apoptosis Inducing Factor genetics, Cell Death drug effects, Conserved Sequence, Humans, Mitochondria metabolism, Apoptosis Inducing Factor metabolism, Caspases metabolism, Cell Death physiology, Molecular Targeted Therapy
- Abstract
Cell death has been initially divided into apoptosis, in which the cell plays an active role, and necrosis, which is considered a passive cell death program. Intense research performed in the last decades has concluded that "programmed" cell death (PCD) is a more complex physiological process than initially thought. Indeed, although in most cases the PCD process is achieved via a family of Cys proteases known as caspases, an important number of regulated PCD pathways do not involve this family of proteases. As a consequence, active forms of PCD are initially referred to as caspase-dependent and caspase-independent. More recent data has revealed that there are also active caspase-independent necrotic pathways defined as necroptosis (programmed necrosis). The existence of necroptotic forms of death was corroborated by the discovery of key executioners such as the kinase RIP1 or the mitochondrial protein apoptosis-inducing factor (AIF). AIF is a Janus protein with a redox activity in the mitochondria and a pro-apoptotic function in the nucleus. We have recently described a particular form of AIF-mediated caspase-independent necroptosis that also implicates other molecules such as PARP-1, calpains, Bax, Bcl-2, histone H2AX, and cyclophilin A. From a therapeutic point of view, the unraveling of this new form of PCD poses a question: is it possible to modulate this necroptotic pathway independently of the classical apoptotic paths? Because the answer is yes, a wider understanding of AIF-mediated necroptosis could theoretically pave the way for the development of new drugs that modulate PCD. To this end, we present here an overview of the current knowledge of AIF and AIF-mediated necroptosis. We also summarize the state of the art in some of the most interesting therapeutic strategies that could target AIF or the AIF-mediated necroptotic pathway., (Copyright © 2011 Wiley Periodicals, Inc.)
- Published
- 2011
- Full Text
- View/download PDF
36. Functional assessment of p53 in chronic lymphocytic leukemia.
- Author
-
Le Garff-Tavernier M, Blons H, Nguyen-Khac F, Pannetier M, Brissard M, Gueguen S, Jacob F, Ysebaert L, Susin SA, and Merle-Béral H
- Published
- 2011
- Full Text
- View/download PDF
37. Histone H2AX: The missing link in AIF-mediated caspase-independent programmed necrosis.
- Author
-
Baritaud M, Boujrad H, Lorenzo HK, Krantic S, and Susin SA
- Subjects
- Apoptosis, Apoptosis Inducing Factor chemistry, Calpain metabolism, Cyclophilins chemistry, Histones chemistry, Humans, bcl-2-Associated X Protein metabolism, Apoptosis Inducing Factor metabolism, Caspases metabolism, Histones metabolism, Necrosis metabolism
- Abstract
Caspase-independent programmed necrosis is a highly regulated cellular demise that displays morphological and biochemical necrotic hallmarks, such as an earlier permeability of the plasma membrane and lactate dehydrogenase (LDH) leakiness. This form of programmed cell death (PCD) is regulated by AIF, a FAD-dependent oxidoreductase, which is released from the mitochondria to the nucleus where it induces chromatin tcondensation and DNA fragmentation. Some years ago, it was established that the sequential activation of poly(ADP-ribose) polymerase- 1 (PARP-1), calpains and Bax regulates the mitochondrial AIF release associated to programmed necrosis. But, what happens when AIF is in the nucleus? How does this protein induce chromatinolysis and programmed necrosis? Recently, we have unraveled some of the mechanisms underlying the nuclear action of AIF in this type of caspase-independent cell death. Indeed, AIF plays a key role in programmed necrosis by its ability to organize a DNA-degrading complex with H2AX and Cyclophiline A (CypA). The AIF/H2AX link is indeed a critical event and explains the nuclear AIF apoptogenic action. In the present article, we outline the current knowledge on cell death by programmed necrosis and discuss the relevance of the AIF/H2AX/CypA DNA-degrading complex in the regulation of this original form of cell death.
- Published
- 2010
- Full Text
- View/download PDF
38. AIF promotes chromatinolysis and caspase-independent programmed necrosis by interacting with histone H2AX.
- Author
-
Artus C, Boujrad H, Bouharrour A, Brunelle MN, Hoos S, Yuste VJ, Lenormand P, Rousselle JC, Namane A, England P, Lorenzo HK, and Susin SA
- Subjects
- Animals, Apoptosis Inducing Factor chemistry, Calpain metabolism, Cell Line, Cyclophilin A genetics, Cyclophilin A metabolism, DNA Damage, Down-Regulation, Fibroblasts cytology, Fibroblasts metabolism, Gene Deletion, Histones chemistry, Histones genetics, Methylnitronitrosoguanidine pharmacology, Mice, Models, Molecular, Poly (ADP-Ribose) Polymerase-1, Poly(ADP-ribose) Polymerases metabolism, bcl-2-Associated X Protein metabolism, Apoptosis Inducing Factor metabolism, Caspases metabolism, Chromatin metabolism, Histones metabolism, Necrosis metabolism
- Abstract
Programmed necrosis induced by DNA alkylating agents, such as MNNG, is a caspase-independent mode of cell death mediated by apoptosis-inducing factor (AIF). After poly(ADP-ribose) polymerase 1, calpain, and Bax activation, AIF moves from the mitochondria to the nucleus where it induces chromatinolysis and cell death. The mechanisms underlying the nuclear action of AIF are, however, largely unknown. We show here that, through its C-terminal proline-rich binding domain (PBD, residues 543-559), AIF associates in the nucleus with histone H2AX. This interaction regulates chromatinolysis and programmed necrosis by generating an active DNA-degrading complex with cyclophilin A (CypA). Deletion or directed mutagenesis in the AIF C-terminal PBD abolishes AIF/H2AX interaction and AIF-mediated chromatinolysis. H2AX genetic ablation or CypA downregulation confers resistance to programmed necrosis. AIF fails to induce chromatinolysis in H2AX or CypA-deficient nuclei. We also establish that H2AX is phosphorylated at Ser139 after MNNG treatment and that this phosphorylation is critical for caspase-independent programmed necrosis. Overall, our data shed new light in the mechanisms regulating programmed necrosis, elucidate a key nuclear partner of AIF, and uncover an AIF apoptogenic motif.
- Published
- 2010
- Full Text
- View/download PDF
39. Caspase-independent type III PCD: a new means to modulate cell death in chronic lymphocytic leukemia.
- Author
-
Merle-Béral H, Barbier S, Roué G, Bras M, Sarfati M, and Susin SA
- Subjects
- Adenine analogs & derivatives, Adenine pharmacology, CD47 Antigen metabolism, Cell Death drug effects, Dynamins, GTP Phosphohydrolases metabolism, Humans, Microtubule-Associated Proteins metabolism, Mitochondrial Proteins metabolism, Caspases metabolism, Cell Death physiology, Leukemia, Lymphocytic, Chronic, B-Cell enzymology, Leukemia, Lymphocytic, Chronic, B-Cell pathology
- Published
- 2009
- Full Text
- View/download PDF
40. Caspase-independent type III programmed cell death in chronic lymphocytic leukemia: the key role of the F-actin cytoskeleton.
- Author
-
Barbier S, Chatre L, Bras M, Sancho P, Roué G, Virely C, Yuste VJ, Baudet S, Rubio M, Esquerda JE, Sarfati M, Merle-Béral H, and Susin SA
- Subjects
- B-Lymphocytes pathology, Caspases, Cytoskeleton immunology, Humans, Leukemia, Lymphocytic, Chronic, B-Cell immunology, Leukemia, Lymphocytic, Chronic, B-Cell metabolism, Tumor Cells, Cultured, Actins metabolism, Apoptosis immunology, Cytoskeleton pathology, Leukemia, Lymphocytic, Chronic, B-Cell pathology
- Abstract
Background: Programmed cell death has been traditionally related with caspase activation. However, it is now accepted that caspase-independent forms of programmed cell death also regulate cell death. In chronic lymphocytic leukemia, CD47 ligation induces one of these alternative forms of cell death: type III programmed cell death. This poorly understood process is characterized by cytoplasmic hallmarks, such as mitochondrial damage. To gain insights into the molecular pathways regulating type III programmed cell death in chronic lymphocytic leukemia, we performed extensive biochemical and cell biology assessments., Design and Methods: After CD47 triggering, purified B-cells from 20 patients with chronic lymphocytic leukemia were studied by flow cytometry, immunofluorescence and three-dimensional imaging, immunoblotting, electron microscopy, and fibrillar/globular actin measurements. Finally, we subjected CD47-treated chronic lymphocytic leukemia cells to a phagocytosis assay., Results: We first confirmed that induction of type III programmed cell death is an efficient means of triggering cell death in chronic lymphocytic leukemia. Further, we demonstrated that the signaling events induced by CD47 ligation provoked a reduction in cell size. This alteration is related to F-actin disruption, as the two other cytoskeleton networks, microtubules and intermediate filaments, remain undisturbed in type III programmed cell death. Strikingly, we revealed that the pharmacological modulation of F-actin dynamics regulated this type of death. Finally, our data delineated a new programmed cell death pathway in chronic lymphocytic leukemia initiated by CD47 triggering, and followed by serine protease activation, F-actin rearrangement, mitochondrial damage, phosphatidylserine exposure, and cell clearance., Conclusions: Our work reveals a key molecular tool in the modulation of cell death in chronic lymphocytic leukemia: F-actin. By assessing the regulation of F-actin and type III programmed cell death, this analysis provides new options for destroying chronic lymphocytic leukemia cells, such as a combination of therapies based on apoptosis regulators (e.g., caspases, Bcl-2, Bax) along with alternative therapies based on type III death effectors (e.g., F-actin).
- Published
- 2009
- Full Text
- View/download PDF
41. Highly cytotoxic and neurotoxic acetogenins of the Annonaceae: new putative biological targets of squamocin detected by activity-based protein profiling.
- Author
-
Derbré S, Gil S, Taverna M, Boursier C, Nicolas V, Demey-Thomas E, Vinh J, Susin SA, Hocquemiller R, and Poupon E
- Subjects
- Electrophoresis, Polyacrylamide Gel, Kinetics, Microscopy, Confocal, Mitochondria drug effects, Proteins isolation & purification, Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization, Acetogenins pharmacology, Annonaceae chemistry, Furans pharmacology, Lactones pharmacology, Neurons drug effects, Proteins metabolism
- Abstract
Acetogenins of the Annonaceae are strong inhibitors of mitochondrial complex I but discrepancies in the structure/activity relationships pled the search for other targets within the whole cell proteome. Combining hemisynthetic work, Cu-catalyzed Huisgen cycloaddition and proteomic techniques we have identified new putative protein targets of squamocin ruling out the previously accepted 'complex I dogma'. These results give new insights into the mechanism of action of these potent neurotoxic molecules.
- Published
- 2008
- Full Text
- View/download PDF
42. Therapeutic potential of AIF-mediated caspase-independent programmed cell death.
- Author
-
Lorenzo HK and Susin SA
- Subjects
- Amino Acid Sequence, Animals, Antineoplastic Agents pharmacology, Apoptosis Inducing Factor chemistry, Cardiovascular Agents pharmacology, Cell Nucleus enzymology, Cell Nucleus metabolism, Cell Nucleus pathology, Drug Resistance, Neoplasm, Enzyme Activation, Humans, Ischemia drug therapy, Ischemia metabolism, Ischemia pathology, Mitochondria enzymology, Mitochondria metabolism, Mitochondria pathology, Models, Molecular, Molecular Sequence Data, Neoplasms drug therapy, Neoplasms metabolism, Neoplasms pathology, Neurodegenerative Diseases drug therapy, Neurodegenerative Diseases metabolism, Neurodegenerative Diseases pathology, Neuroprotective Agents pharmacology, Neuroprotective Agents therapeutic use, Protein Conformation, Protein Isoforms metabolism, Antineoplastic Agents therapeutic use, Apoptosis drug effects, Apoptosis Inducing Factor metabolism, Cardiovascular Agents therapeutic use, Caspases metabolism, Cell Nucleus drug effects, Mitochondria drug effects
- Abstract
Resistance to anticancer drugs is often related to deficient cell death execution pathways in cancer cells. Apoptosis, which denotes a form of cell death executed by caspases, was traditionally considered as the only physiological and programmed form of cell death. However, recent evidence indicates that programmed cell death (PCD) can occur in complete absence of caspase activation. Indeed, a large number of caspase-independent models are now defined and a key protein implicated in this type of PCD, apoptosis-inducing factor (AIF), has been identified. AIF is a mitochondrial protein with two faces looking in opposite life/death directions. Recently, the identification of five different isoforms allowed a better characterization of AIFs life/mitochondrial versus death/nuclear functions, as well as definition of its pro-apoptotic region and some of its nuclear partners. Importantly, much work on caspase-independent PCD has revealed that AIF participates in more PCD systems than initially thought. A wider molecular knowledge of AIF, and of the caspase-independent PCDs in which it is involved, are key to provide new insights into the role of PCD. There is no doubt that these insights will lead to the development of more selective and efficient drugs against cancer, degenerative diseases, and other pathological disorders implicating AIF.
- Published
- 2007
- Full Text
- View/download PDF
43. AIF-mediated programmed necrosis: a highly regulated way to die.
- Author
-
Boujrad H, Gubkina O, Robert N, Krantic S, and Susin SA
- Subjects
- Animals, Apoptosis genetics, Apoptosis physiology, Cell Death genetics, Cell Death physiology, Humans, Necrosis classification, Necrosis enzymology, Necrosis genetics, Necrosis pathology, Apoptosis Inducing Factor physiology
- Abstract
Historically, two main forms of cell death have been distinguished: apoptosis and necrosis. Apoptosis was initially considered as the only physiological and programmed form of cell death. This type of death is recurrently associated with caspases, a family of cysteine proteases activated in apoptotic conditions. However, it is now widely recognized that programmed cell death (PCD) can also occur in the complete absence of caspase activation. The existence of non-caspase PCD pathways was corroborated by the discovery of caspase-independent executioners, such as the mitochondrial protein Apoptosis-Inducing Factor (AIF). Necrosis has often been viewed as an accidental and uncontrolled cell death process. Nevertheless, increasing evidence shows that, like apoptosis, necrosis could be a highly orchestrated type of PCD. Indeed, apoptosis and necrosis present more similarities than it has been originally thought. Here, we summarize the different classifications of PCD and the current knowledge of a necrotic PCD pathway mediated by AIF: alkylating DNA-damage mediated death. We also outline the molecular mechanisms controlling this form of PCD and discuss their potential relevance in physiological and pathological settings. These emerging data on the molecular mechanisms regulating programmed necrosis may certainly have potent therapeutic consequences in treating both apoptotic-resistant tumors and degenerating adult neurons.
- Published
- 2007
- Full Text
- View/download PDF
44. Drp1 mediates caspase-independent type III cell death in normal and leukemic cells.
- Author
-
Bras M, Yuste VJ, Roué G, Barbier S, Sancho P, Virely C, Rubio M, Baudet S, Esquerda JE, Merle-Béral H, Sarfati M, and Susin SA
- Subjects
- Active Transport, Cell Nucleus physiology, CD47 Antigen genetics, CD47 Antigen metabolism, Cell Shape, Cells, Cultured, Dynamins, Electron Transport physiology, GTP Phosphohydrolases genetics, Humans, Microtubule-Associated Proteins genetics, Mitochondria metabolism, Mitochondria pathology, Mitochondrial Proteins genetics, Proto-Oncogene Proteins c-bcl-2 genetics, Proto-Oncogene Proteins c-bcl-2 metabolism, Serine Endopeptidases metabolism, T-Lymphocytes cytology, T-Lymphocytes physiology, Caspases metabolism, Cell Death physiology, GTP Phosphohydrolases metabolism, Leukemia metabolism, Microtubule-Associated Proteins metabolism, Mitochondrial Proteins metabolism
- Abstract
Ligation of CD47 triggers caspase-independent programmed cell death (PCD) in normal and leukemic cells. Here, we characterize the morphological and biochemical features of this type of death and show that it displays the hallmarks of type III PCD. A molecular and biochemical approach has led us to identify a key mediator of this type of death, dynamin-related protein 1 (Drp1). CD47 ligation induces Drp1 translocation from cytosol to mitochondria, a process controlled by chymotrypsin-like serine proteases. Once in mitochondria, Drp1 provokes an impairment of the mitochondrial electron transport chain, which results in dissipation of mitochondrial transmembrane potential, reactive oxygen species generation, and a drop in ATP levels. Surprisingly, neither the activation of the most representative proapoptotic members of the Bcl-2 family, such as Bax or Bak, nor the release of apoptogenic proteins AIF (apoptosis-inducing factor), cytochrome c, endonuclease G (EndoG), Omi/HtrA2, or Smac/DIABLO from mitochondria to cytosol is observed. Responsiveness of cells to CD47 ligation increases following Drp1 overexpression, while Drp1 downregulation confers resistance to CD47-mediated death. Importantly, in B-cell chronic lymphocytic leukemia cells, mRNA levels of Drp1 strongly correlate with death sensitivity. Thus, this previously unknown mechanism controlling caspase-independent type III PCD may provide the basis for novel therapeutic approaches to overcome apoptotic avoidance in malignant cells.
- Published
- 2007
- Full Text
- View/download PDF
45. Sequential activation of poly(ADP-ribose) polymerase 1, calpains, and Bax is essential in apoptosis-inducing factor-mediated programmed necrosis.
- Author
-
Moubarak RS, Yuste VJ, Artus C, Bouharrour A, Greer PA, Menissier-de Murcia J, and Susin SA
- Subjects
- Alkylating Agents pharmacology, Alkylation drug effects, Animals, Caspases metabolism, Cell Death drug effects, Cell Nucleus drug effects, Cell Nucleus metabolism, Cytosol drug effects, Cytosol metabolism, DNA metabolism, Enzyme Activation drug effects, Fibroblasts cytology, Fibroblasts drug effects, Fibroblasts enzymology, Methylnitronitrosoguanidine pharmacology, Mice, Mitochondria drug effects, Mitochondria metabolism, Models, Biological, Protein Transport drug effects, Tumor Suppressor Protein p53 metabolism, bcl-2 Homologous Antagonist-Killer Protein metabolism, Apoptosis Inducing Factor metabolism, Calpain metabolism, Necrosis pathology, Poly(ADP-ribose) Polymerases metabolism, bcl-2-Associated X Protein metabolism
- Abstract
Alkylating DNA damage induces a necrotic type of programmed cell death through the poly(ADP-ribose) polymerases (PARP) and apoptosis-inducing factor (AIF). Following PARP activation, AIF is released from mitochondria and translocates to the nucleus, where it causes chromatin condensation and DNA fragmentation. By employing a large panel of gene knockout cells, we identified and describe here two essential molecular links between PARP and AIF: calpains and Bax. Alkylating DNA damage initiated a p53-independent form of death involving PARP-1 but not PARP-2. Once activated, PARP-1 mediated mitochondrial AIF release and necrosis through a mechanism requiring calpains but not cathepsins or caspases. Importantly, single ablation of the proapoptotic Bcl-2 family member Bax, but not Bak, prevented both AIF release and alkylating DNA damage-induced death. Thus, Bax is indispensable for this type of necrosis. Our data also revealed that Bcl-2 regulates N-methyl-N'-nitro-N'-nitrosoguanidine-induced necrosis. Finally, we established the molecular ordering of PARP-1, calpains, Bax, and AIF activation, and we showed that AIF downregulation confers resistance to alkylating DNA damage-induced necrosis. Our data shed new light on the mechanisms regulating AIF-dependent necrosis and support the notion that, like apoptosis, necrosis could be a highly regulated cell death program.
- Published
- 2007
- Full Text
- View/download PDF
46. Expression of cortical and hippocampal apoptosis-inducing factor (AIF) in aging and Alzheimer's disease.
- Author
-
Reix S, Mechawar N, Susin SA, Quirion R, and Krantic S
- Subjects
- Adult, Age Factors, Aged, Aged, 80 and over, Aging metabolism, Alzheimer Disease metabolism, Analysis of Variance, Blotting, Western methods, Female, Humans, Male, Middle Aged, Aging pathology, Alzheimer Disease pathology, Apoptosis Inducing Factor metabolism, Cerebral Cortex metabolism, Hippocampus metabolism, Up-Regulation physiology
- Abstract
Apoptosis-inducing factor (AIF) is a mitochondrial oxidoreductase originally identified for its role in caspase-independent programmed cell death (PCD). In this study, we investigated AIF protein expression levels in frontal and temporal cortices of normal subjects of various ages, as well as in subjects with Alzheimer's disease (AD). AIF levels were also measured in the hippocampus of age-matched elderly and AD subjects. Amounts of all three AIF isoforms increased significantly with age in both cortical areas. Interestingly, AIF expression levels in the cortex (but not hippocampus) were consistently lower in AD compared to age-matched controls. The up-regulation of cortical AIF in normal aging is consistent with its previously hypothesized role as a free radical scavenger, and may thus represent an adaptive cellular response to compensate for the steady increase in oxidative stress occurring with age.
- Published
- 2007
- Full Text
- View/download PDF
47. CD44 ligation induces caspase-independent cell death via a novel calpain/AIF pathway in human erythroleukemia cells.
- Author
-
Artus C, Maquarre E, Moubarak RS, Delettre C, Jasmin C, Susin SA, and Robert-Lézénès J
- Subjects
- Antigens, CD physiology, Apoptosis, Apoptosis Inducing Factor antagonists & inhibitors, Apoptosis Inducing Factor genetics, Caspases metabolism, Cell Cycle, Cell Line, Tumor, Cell Survival, Cytochromes c analysis, DNA, Neoplasm isolation & purification, Electrophoresis, Gel, Pulsed-Field, Humans, Isoquinolines pharmacology, Membrane Potentials, Mitochondrial Membranes physiology, Poly(ADP-ribose) Polymerase Inhibitors, Protein Transport, RNA, Small Interfering genetics, Transfection, Apoptosis Inducing Factor metabolism, Calpain metabolism, Cell Death physiology, Hyaluronan Receptors physiology, Leukemia, Erythroblastic, Acute pathology
- Abstract
Ligation of the cell surface molecule CD44 by anti-CD44 monoclonal antibodies (mAbs) has been shown to induce cell differentiation, cell growth inhibition and in some cases, apoptosis in myeloid leukemic cells. We report, herein, that exposure of human erythroleukemic HEL cells to the anti-CD44 mAb A3D8 resulted in cell growth inhibition followed by caspase-independent apoptosis-like cell death. This process was associated with the disruption of mitochondrial membrane potential (Delta Psi m), the mitochondrial release of apoptosis-inducing factor (AIF), but not of cytochrome c, and the nuclear translocation of AIF. All these effects including cell death, loss of mitochondrial Delta Psi m and AIF release were blocked by pretreatment with the poly (ADP-ribose) polymerase inhibitor isoquinoline. A significant protection against cell death was also observed by using small interfering RNA for AIF. Moreover, we show that calpain protease was activated before the appearance of apoptosis, and that calpain inhibitors or transfection of calpain-siRNA decrease A3D8-induced cell death, and block AIF release. These data suggest that CD44 ligation triggers a novel caspase-independent cell death pathway via calpain-dependent AIF release in erythroleukemic HEL cells.
- Published
- 2006
- Full Text
- View/download PDF
48. Identification and characterization of AIFsh2, a mitochondrial apoptosis-inducing factor (AIF) isoform with NADH oxidase activity.
- Author
-
Delettre C, Yuste VJ, Moubarak RS, Bras M, Robert N, and Susin SA
- Subjects
- Alternative Splicing, Amino Acid Sequence, Animals, Apoptosis, Apoptosis Inducing Factor metabolism, HeLa Cells, Humans, Isoenzymes genetics, Isoenzymes metabolism, Mice, Mitochondria, Liver enzymology, Molecular Sequence Data, Multienzyme Complexes genetics, NADH, NADPH Oxidoreductases genetics, Organ Specificity, RNA, Messenger genetics, Reactive Oxygen Species metabolism, Sequence Alignment, Transcription, Genetic, Apoptosis Inducing Factor genetics, Exons genetics, Mitochondria enzymology
- Abstract
Apoptosis-inducing factor (AIF) is a bifunctional NADH oxidase involved in mitochondrial respiration and caspase-independent apoptosis. Three alternatively spliced mRNA isoforms of AIF have been identified previously: AIF, AIF-exB, and AIFsh. Here, we report the cloning and the biochemical characterization of a new isoform named AIF short 2 (AIFsh2). AIFsh2 transcript includes a previously unknown exon placed between exons 9 and 10 of AIF. The resulting AIFsh2 protein, which localizes in mitochondria, corresponds to the oxidoreductase domain of AIF. In this way, AIFsh2 exhibits similar NADH oxidase activity to AIF and generates reactive oxygen species. Like AIF, AIFsh2 is released from mitochondria to cytosol after an apoptotic insult in a calpain or cathepsin-dependent manner. However, in contrast to AIF, AIFsh2 does not induce nuclear apoptosis. Thus, it seems that the reactive oxygen species produced by the oxidoreductase domain of AIF/AIFsh2 are not important for AIF-dependent nuclear apoptosis. In addition, we demonstrate that the AIFsh2 mRNA is absent in normal brain tissue, whereas it is expressed in neuroblastoma-derived cells, suggesting a different regulation in normal and transformed cells from the brain lineage. Together, our results reveal that AIF yields an original and independent genetic regulation of the two AIF functions. This is an important issue to understand the physiological role of this protein.
- Published
- 2006
- Full Text
- View/download PDF
49. Regulation of apoptosis/necrosis execution in cadmium-treated human promonocytic cells under different forms of oxidative stress.
- Author
-
Sancho P, Fernández C, Yuste VJ, Amrán D, Ramos AM, de Blas E, Susin SA, and Aller P
- Subjects
- Acetylcysteine pharmacology, Adenosine Triphosphate analysis, Adenosine Triphosphate metabolism, Antimetabolites, Antineoplastic pharmacology, Benzamides pharmacology, Buthionine Sulfoximine pharmacology, Cadmium analysis, Caspase 3, Caspase 9, Caspases metabolism, Drug Interactions, Glutathione analysis, Glutathione metabolism, Humans, Hydrogen Peroxide pharmacology, Membrane Potentials drug effects, Mitochondria drug effects, Necrosis chemically induced, Necrosis pathology, Oxidants pharmacology, Proto-Oncogene Proteins c-bcl-2 metabolism, RNA, Small Interfering metabolism, Transfection, U937 Cells, Apoptosis drug effects, Cadmium pharmacology, Monocytes drug effects, Oxidative Stress
- Abstract
Pulse-treatment of U-937 human promonocytic cells with cadmium chloride followed by recovery caused caspase-9/caspase-3-dependent, caspase-8-independent apoptosis. However, pre-incubation with the glutathione (GSH)-suppressing agent DL-buthionine-(S,R)-sulfoximine (cadmium/BSO), or co-treatment with H2O2 (cadmium/H2O2), switched the mode of death to caspase-independent necrosis. The switch from apoptosis to necrosis did not involve gross alterations in Apaf-1 and pro-caspase-9 expression, nor inhibition of cytochrome c release from mitochondria. However, cadmium/H2O2-induced necrosis involved ATP depletion and was prevented by 3-aminobenzamide, while cadmium/BSO-induced necrosis was ATP independent. Pre-incubation with BSO increased the intracellular cadmium accumulation, while co-treatment with H2O2 did not. Both treatments caused intracellular peroxide over-accumulation and disruption of mitochondrial transmembrane potential (delta psi m). However, while post-treatment with N-acetyl-L-cysteine or butylated hydroxyanisole reduced the cadmium/BSO-mediated necrosis and delta psi m disruption, it did not reduce the effects of cadmium/H2O2. Bcl-2 over-expression, which reduced peroxide accumulation without affecting the intracellular GSH content, attenuated necrosis generation by cadmium/H2O2 but not by cadmium/BSO. By contrast, AIF suppression, which reduced peroxide accumulation and increased the GSH content, attenuated the toxicity of both treatments. These results unravel the existence of two different oxidation-mediated necrotic pathways in cadmium-treated cells, one of them resulting from ATP-dependent apoptosis blockade, and the other involving the concurrence of multiple regulatory factors.
- Published
- 2006
- Full Text
- View/download PDF
50. Use of penetrating peptides interacting with PP1/PP2A proteins as a general approach for a drug phosphatase technology.
- Author
-
Guergnon J, Dessauge F, Dominguez V, Viallet J, Bonnefoy S, Yuste VJ, Mercereau-Puijalon O, Cayla X, Rebollo A, Susin SA, Bost PE, and Garcia A
- Subjects
- Amino Acid Sequence, Animals, Casein Kinase II metabolism, Catalytic Domain, Chick Embryo, HeLa Cells, Humans, Jurkat Cells, Molecular Sequence Data, Peptides chemistry, Peptides metabolism, Phosphoprotein Phosphatases chemistry, Protein Phosphatase 1, Sequence Homology, Amino Acid, Phosphoprotein Phosphatases metabolism
- Abstract
Protein phosphatase types 1 (PP1) and 2A (PP2A) represent two major families of serine/threonine protein phosphatases that have been implicated in the regulation of many cellular processes, including cell growth and apoptosis in mammalian cells. PP1 and PP2A proteins are composed of oligomeric complexes comprising a catalytic structure (PP1c or PP2AC) containing the enzymatic activity and at least one more interacting subunit. The binding of different subunits to a catalytic structure generates a broad variety of holoenzymes. We showed here that casein kinase 2alpha (Ck2alpha) and simian virus 40 small t antigen share a putative common beta-strand structure required for PP2A1 trimeric holoenzyme binding. We have also characterized DPT-sh1, a short basic peptide from Ck2alpha that interacted only in vitro with the PP2A-A subunit and behaves as a nontoxic penetrating shuttle in several cultivated human cell lines and chick embryos. In addition, DPT-sh1 specifically accumulated in human red cells infected with Plasmodium falciparum malaria parasites. We therefore designed bipartite peptides containing DPT-sh1 and PP1- or PP2A-interacting sequences. We found that DPT-5, a DPT-sh1-derived peptide containing a short sequence identified in CD28 antigen, interacts with PP2A-Balpha, and DPT-7, another DPT-sh1-derived peptide containing a short sequence identified in Bad as a PP1 catalytic consensus docking motif, induce apoptosis in cultivated cell lines. These results clearly indicate that the rational design of PP1/PP2A interacting peptides is a pertinent strategy to deregulate intracellular survival pathways.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.