Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Cell Death Differ ; 31(3): 335-347, 2024 03.
Article in English | MEDLINE | ID: mdl-38396150

ABSTRACT

During apoptosis mediated by the intrinsic pathway, BAX/BAK triggers mitochondrial permeabilization and the release of cytochrome-c, followed by a dramatic remodelling of the mitochondrial network that results in mitochondrial herniation and the subsequent release of pro-inflammatory mitochondrial components. Here, we show that mitochondrial herniation and subsequent exposure of the inner mitochondrial membrane (IMM) to the cytoplasm, initiates a unique form of mitophagy to deliver these damaged organelles to lysosomes. IMM-induced mitophagy occurs independently of canonical PINK1/Parkin signalling and is driven by ubiquitination of the IMM. Our data suggest IMM-induced mitophagy is an additional safety mechanism that cells can deploy to contain damaged mitochondria. It may have particular relevance in situations where caspase activation is incomplete or inhibited, and in contexts where PINK1/Parkin-mitophagy is impaired or overwhelmed.


Subject(s)
Mitophagy , Ubiquitin-Protein Ligases , Humans , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Mitochondrial Membranes/metabolism , Protein Kinases/metabolism
2.
Cell Rep ; 36(3): 109430, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34289356

ABSTRACT

While the intrinsic apoptosis pathway is thought to play a central role in shaping the B cell lineage, its precise role in mature B cell homeostasis remains elusive. Using mice in which mature B cells are unable to undergo apoptotic cell death, we show that apoptosis constrains follicular B (FoB) cell lifespan but plays no role in marginal zone B (MZB) cell homeostasis. In these mice, FoB cells accumulate abnormally. This intensifies intercellular competition for BAFF, resulting in a contraction of the MZB cell compartment, and reducing the growth, trafficking, and fitness of FoB cells. Diminished BAFF signaling dampens the non-canonical NF-κB pathway, undermining FoB cell growth despite the concurrent triggering of a protective p53 response. Thus, MZB and FoB cells exhibit a differential requirement for the intrinsic apoptosis pathway. Homeostatic apoptosis constrains the size of the FoB cell compartment, thereby preventing competition-induced FoB cell atrophy.


Subject(s)
Apoptosis , B-Lymphocytes/pathology , Homeostasis , Animals , Antibody Formation/immunology , Atrophy , B-Cell Activating Factor/metabolism , Cell Count , Cell Differentiation/genetics , Cell Proliferation/genetics , Cell Size , Cell Survival/genetics , Cellular Senescence/genetics , Gene Deletion , Gene Expression Regulation , Mice, Knockout , Sequence Analysis, RNA , Thymus Gland/immunology , Transcription Factors/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
3.
Cell Rep ; 31(1): 107492, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32268090

ABSTRACT

Stimulator of Interferon Genes (STING) is a critical component of host innate immune defense but can contribute to chronic autoimmune or autoinflammatory disease. Once activated, the cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) (cGAMP) synthase (cGAS)-STING pathway induces both type I interferon (IFN) expression and nuclear factor-κB (NF-κB)-mediated cytokine production. Currently, these two signaling arms are thought to be mediated by a single upstream kinase, TANK-binding kinase 1 (TBK1). Here, using genetic and pharmacological approaches, we show that TBK1 alone is dispensable for STING-induced NF-κB responses in human and mouse immune cells, as well as in vivo. We further demonstrate that TBK1 acts redundantly with IκB kinase ε (IKKε) to drive NF-κB upon STING activation. Interestingly, we show that activation of IFN regulatory factor 3 (IRF3) is highly dependent on TBK1 kinase activity, whereas NF-κB is significantly less sensitive to TBK1/IKKε kinase inhibition. Our work redefines signaling events downstream of cGAS-STING. Our findings further suggest that cGAS-STING will need to be targeted directly to effectively ameliorate the inflammation underpinning disorders associated with STING hyperactivity.


Subject(s)
I-kappa B Kinase/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Female , HEK293 Cells , Humans , I-kappa B Kinase/physiology , Immunity, Innate , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Male , Membrane Proteins/metabolism , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , NF-kappa B/metabolism , Nucleotides, Cyclic/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/physiology , Signal Transduction/immunology
4.
Cell Death Differ ; 26(9): 1766-1781, 2019 09.
Article in English | MEDLINE | ID: mdl-30538285

ABSTRACT

Drugs targeting various pro-survival BCL-2 family members (''BH3 mimetics'') have efficacy in hemopoietic malignancies, but the non-targeted pro-survival family members can promote resistance. Pertinently, the sensitivity of some tumor cell lines to BH3 mimetic ABT737, which targets BCL-2, BCL-XL, and BCL-W but not MCL-1, is enhanced by 2-deoxyglucose (2DG). We found that 2DG augmented apoptosis induced by ABT737 in 3 of 8 human hemopoietic tumor cell lines, most strongly in pre-B acute lymphocytic leukemia cell line NALM-6, the focus of our mechanistic studies. Although 2DG can lower MCL-1 translation, how it does so is incompletely understood, in part because 2DG inhibits both glycolysis and protein glycosylation in the endoplasmic reticulum (ER). Its glycolysis inhibition lowered ATP and, through the AMPK/mTORC1 pathway, markedly reduced global protein synthesis, as did an ER integrated stress response. A dual reporter assay revealed that 2DG impeded not only cap-dependent translation but also elongation or cap-independent translation. MCL-1 protein fell markedly, whereas 12 other BCL-2 family members were unaffected. We ascribe the MCL-1 drop to the global fall in translation, exacerbated for mRNAs with a structured 5' untranslated region (5'UTR) containing potential regulatory motifs like those in MCL-1 mRNA and the short half-life of MCL-1 protein. Pertinently, 2DG downregulated two other short-lived oncoproteins, MYC and MDM2. Thus, our results support MCL-1 as a critical 2DG target, but also reveal multiple effects on global translation that may well also affect its promotion of apoptosis.


Subject(s)
Myeloid Cell Leukemia Sequence 1 Protein/genetics , Peptide Fragments/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Protein Biosynthesis , Proto-Oncogene Proteins/genetics , AMP-Activated Protein Kinase Kinases , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Biomimetics , Biphenyl Compounds , Cell Line, Tumor , Deoxyglucose/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Kinases/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/genetics
7.
Science ; 337(6102): 1678-84, 2012 Sep 28.
Article in English | MEDLINE | ID: mdl-23019653

ABSTRACT

Cancer cells accommodate multiple genetic and epigenetic alterations that initially activate intrinsic (cell-autonomous) and extrinsic (immune-mediated) oncosuppressive mechanisms. Only once these barriers to oncogenesis have been overcome can malignant growth proceed unrestrained. Tetraploidization can contribute to oncogenesis because hyperploid cells are genomically unstable. We report that hyperploid cancer cells become immunogenic because of a constitutive endoplasmic reticulum stress response resulting in the aberrant cell surface exposure of calreticulin. Hyperploid, calreticulin-exposing cancer cells readily proliferated in immunodeficient mice and conserved their increased DNA content. In contrast, hyperploid cells injected into immunocompetent mice generated tumors only after a delay, and such tumors exhibited reduced DNA content, endoplasmic reticulum stress, and calreticulin exposure. Our results unveil an immunosurveillance system that imposes immunoselection against hyperploidy in carcinogen- and oncogene-induced cancers.


Subject(s)
Endoplasmic Reticulum Stress/immunology , Immunologic Surveillance , Neoplasms/genetics , Neoplasms/immunology , Ploidies , Animals , Calreticulin/immunology , Cell Line, Tumor , Common Variable Immunodeficiency/genetics , DNA, Neoplasm/analysis , DNA, Neoplasm/genetics , Eukaryotic Initiation Factor-2/metabolism , Humans , Immunocompetence , Mice , Mice, Inbred BALB C , Neoplasms/chemically induced , Phosphorylation
8.
Cell Rep ; 2(2): 257-69, 2012 Aug 30.
Article in English | MEDLINE | ID: mdl-22854025

ABSTRACT

Patients with non-small cell lung cancer (NSCLC) are routinely treated with cytotoxic agents such as cisplatin. Through a genome-wide siRNA-based screen, we identified vitamin B6 metabolism as a central regulator of cisplatin responses in vitro and in vivo. By aggravating a bioenergetic catastrophe that involves the depletion of intracellular glutathione, vitamin B6 exacerbates cisplatin-mediated DNA damage, thus sensitizing a large panel of cancer cell lines to apoptosis. Moreover, vitamin B6 sensitizes cancer cells to apoptosis induction by distinct types of physical and chemical stress, including multiple chemotherapeutics. This effect requires pyridoxal kinase (PDXK), the enzyme that generates the bioactive form of vitamin B6. In line with a general role of vitamin B6 in stress responses, low PDXK expression levels were found to be associated with poor disease outcome in two independent cohorts of patients with NSCLC. These results indicate that PDXK expression levels constitute a biomarker for risk stratification among patients with NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Vitamin B 6/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cisplatin/administration & dosage , Cohort Studies , Disease-Free Survival , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Genome-Wide Association Study , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Middle Aged , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Pyridoxal Kinase/biosynthesis , Pyridoxal Kinase/genetics , Survival Rate , Vitamin B 6/genetics
9.
EMBO Mol Med ; 4(6): 500-14, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22438244

ABSTRACT

The genetic or functional inactivation of p53 is highly prevalent in human cancers. Using high-content videomicroscopy based on fluorescent TP53(+/+) and TP53(-/-) human colon carcinoma cells, we discovered that SP600125, a broad-spectrum serine/threonine kinase inhibitor, kills p53-deficient cells more efficiently than their p53-proficient counterparts, in vitro. Similar observations were obtained in vivo, in mice carrying p53-deficient and -proficient human xenografts. Such a preferential cytotoxicity could be attributed to the failure of p53-deficient cells to undergo cell cycle arrest in response to SP600125. TP53(-/-) (but not TP53(+/+) ) cells treated with SP600125 became polyploid upon mitotic abortion and progressively succumbed to mitochondrial apoptosis. The expression of an SP600125-resistant variant of the mitotic kinase MPS1 in TP53(-/-) cells reduced SP600125-induced polyploidization. Thus, by targeting MPS1, SP600125 triggers a polyploidization program that cannot be sustained by TP53(-/-) cells, resulting in the activation of mitotic catastrophe, an oncosuppressive mechanism for the eradication of mitosis-incompetent cells.


Subject(s)
Anthracenes/pharmacology , Antineoplastic Agents/pharmacology , Tumor Suppressor Protein p53/deficiency , Animals , Anthracenes/therapeutic use , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Mice , Microscopy, Video/methods , Neoplasms/drug therapy , Neoplasms/pathology , Xenograft Model Antitumor Assays
10.
Science ; 334(6062): 1573-7, 2011 Dec 16.
Article in English | MEDLINE | ID: mdl-22174255

ABSTRACT

Antineoplastic chemotherapies are particularly efficient when they elicit immunogenic cell death, thus provoking an anticancer immune response. Here we demonstrate that autophagy, which is often disabled in cancer, is dispensable for chemotherapy-induced cell death but required for its immunogenicity. In response to chemotherapy, autophagy-competent, but not autophagy-deficient, cancers attracted dendritic cells and T lymphocytes into the tumor bed. Suppression of autophagy inhibited the release of adenosine triphosphate (ATP) from dying tumor cells. Conversely, inhibition of extracellular ATP-degrading enzymes increased pericellular ATP in autophagy-deficient tumors, reestablished the recruitment of immune cells, and restored chemotherapeutic responses but only in immunocompetent hosts. Thus, autophagy is essential for the immunogenic release of ATP from dying cells, and increased extracellular ATP concentrations improve the efficacy of antineoplastic chemotherapies when autophagy is disabled.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/physiology , Neoplasms/immunology , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Calreticulin/pharmacology , Cell Death/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitoxantrone/pharmacology , Neoplasms/drug therapy
11.
Cell Cycle ; 10(18): 3168-75, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21897118

ABSTRACT

Tyrosine kinases such as SRC family kinases (SFKs) as well as the mammalian target of rapamycin (mTOR) serine/threonine kinase are often constitutively activated in acute myeloid leukemia (AML) and hence constitute potential therapeutic targets. Here we demonstrate that the epidermal growth factor receptor (EGFR) inhibitor erlotinib, which has previously been shown to mediate antiproliferative/cytotoxic off-target effects in myelodysplastic syndrome (MDS) and AML blasts, reduces SFK overactivation. Erlotinib induced an arrest in the G 1 phase of the cell cycle that, in cells with constitutive SFK activation, could be recapitulated by chemical inhibition of SFKs with 3-(4-chlorophenyl)1-(1,1-dimethylethyl)-1H-pyrazolo[3,4-α]pyrimidin-4-amine (PP2). Moreover, erlotinib inhibited the phosphorylation of mTOR targets like p70 (SK6) , stimulated the maturation of the autophagic marker LC3 and promoted the formation of autophagosomes. Notably, PP2 and the mTOR inhibitor rapamycin had a similar cell cycle-arresting activity to erlotinib, but neither of these compounds alone induced significant levels of cell death. Altogether, these results suggest that the therapeutic off-target effect of erlotinib may be linked to, yet cannot be entirely explained by, the inhibition of oncogenic signaling via SFKs and mTOR. Thus, combination therapies with erlotinib and rapamycin might be beneficial for MDS and AML patients.


Subject(s)
Leukemia, Myeloid, Acute/pathology , Quinazolines/pharmacology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , src-Family Kinases/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis , Autophagy , Biomarkers, Tumor/metabolism , Enzyme Activation , Erlotinib Hydrochloride , Flow Cytometry/methods , G1 Phase Cell Cycle Checkpoints , HL-60 Cells/drug effects , Humans , Leukemia, Myeloid, Acute/metabolism , Microscopy, Electron , Microscopy, Fluorescence , Microtubule-Associated Proteins/metabolism , Phosphorylation , Pyrimidines/pharmacology , Signal Transduction , Stilbenes/pharmacology
12.
Biochem Pharmacol ; 82(10): 1457-66, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21664897

ABSTRACT

Initially, tyrosine kinase inhibitors (TKIs) were developed as targeted therapies that would solely interfere with aberrant tyrosine kinase activation in malignant cells. Nevertheless, preclinical and clinical studies demonstrated that TKI also exhibit "off-target" effects, that is effects not mediated by the assumed mechanisms of action. We and others showed that the epidermal growth factor receptor (EGFR) inhibitors erlotinib and gefitinib exert potent antineoplastic effects on EGFR-negative myeloblasts from patients with myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML). Here, we undertook a side-by-side comparison of the anti-leukemic efficacy of four different TKI in MDS and AML. Besides the EGFR inhibitor erlotinib, which served as a point of reference, we employed the dual EGFR/HER2 TKI lapatinib, as well as the multikinase inhibitors dasatinib and sorafenib. All four drugs had anti-leukemic effects on cell line models of MDS/AML in vitro as well as on malignant blasts from MDS/AML patients ex vivo. We explored the biological phenomena underlying this anti-leukemic efficacy. Since it is established that a therapeutic benefit in MDS/AML can be conveyed by induction of cell cycle arrest, apoptosis and/or differentiation, we deciphered the individual contribution of these three phenomena to the anti-leukemic action of each of the four TKI. The concomitant assessment of the panel of TKI enables us thus to define (and quantify) their differential capacity to impact on the three biological phenomena, and provide further evidence that these mechanisms are not solely explained by on-target effects.


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Apoptosis/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Female , Humans , Leukocytes/cytology , Leukocytes/drug effects , Male , Middle Aged
13.
Cell Cycle ; 10(14): 2323-30, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21654193

ABSTRACT

The deregulation of the DNA damage response (DDR) can contribute to leukemogenesis and favor the progression from myelodysplastic syndrome (MDS) to acute myeloid leukemia (AML). Since hypomethylating agent, notably azacitidine, constitute an efficient therapy for patients with high-risk MDS, we assessed whether such compounds can activate the DDR in malignant blasts. While azacitidine and decitabine had moderate effects on apoptosis and cell cycle progression, both agents induced profound changes in the expression and functionality of DDR-related proteins. Decitabine, and to a lesser degree azacitidine, induced the activation of checkpoint kinases Chk-1 and Chk-2 and the phosphorylation of the DDR-sensor H2AX. In addition, hypomethylating agents were found to cause the dephosphorylation of the transcriptional regulator forkhead box O3, best known as FOXO3A, whose phosphorylation has been related to poor prognosis in AML. The dephoasphorylation of FOXO3A induced by azacitidine or decitabine in malignant blasts was accompanied by the translocation of FOXO3A from the cytoplasm to the nucleus. Upon stimulation with azacitidine, MDS/AML-derived, azacitidine-sensitive SKM-1S cells upregulated FOXO3A and the pro-apoptotic FOXO3A targets BIM and PUMA, and this effect was attenuated or abolished in azacitidine-resistant SMK-1R cells. Altogether, our results suggest that the reactivation of FOXO3A may contribute to the effects of hypomethylating agents in malignant blasts.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Forkhead Transcription Factors/metabolism , Leukemia, Myeloid, Acute/metabolism , Adult , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/therapeutic use , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Azacitidine/therapeutic use , Bcl-2-Like Protein 11 , Cell Line, Tumor , Checkpoint Kinase 1 , Checkpoint Kinase 2 , DNA Methylation , DNA Repair , Decitabine , Female , Forkhead Box Protein O3 , Histones/metabolism , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Male , Membrane Proteins/metabolism , Middle Aged , Phosphorylation , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism
14.
Autophagy ; 7(8): 883-91, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21460612

ABSTRACT

Autophagic flux can be measured by determining the declining abundance of autophagic substrates such as sequestosome 1 (SQSTM1, better known as p62), which is sequestered in autophagosomes upon its direct interaction with LC3. However, the total amount of p62 results from two opposed processes, namely its synthesis (which can be modulated by some cellular stressors including autophagy inducers) and its degradation. To avoid this problem, we generated a stable cell line expressing a chimeric protein composed by p62 and the HaloTag (®) protein, which serves as a receptor for fluorescent HaloTag (®) ligands. Upon labeling with HaloTag (®) ligands (which form covalent, near-to-undissociable bonds with the Halotag (®) receptor) and washing, the resulting fluorescent labeling is not influenced by de novo protein synthesis, therefore allowing for the specific monitoring of the fusion protein decline without any interference by protein synthesis. We demonstrate that a HaloTag (®) -p62 fusion protein stably expressed in suitable cell lines can be used to monitor autophagy by flow cytometry and automated fluorescence microscopy. We surmise that this system could be adapted to high-throughput applications.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , Flow Cytometry/methods , Microscopy, Fluorescence/methods , Cell Line, Tumor , Humans , Organelles/metabolism , Subcellular Fractions/metabolism , Substrate Specificity
15.
Autophagy ; 6(1): 189-91, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20110771

ABSTRACT

Cells respond to stress by activating cytoplasmic mechanisms as well as transcriptional programs that can lead to adaptation or death. Autophagy represents an important cytoprotective response that is regulated by both transcriptional and transcription-independent pathways. NFkappaB is perhaps the transcription factor most frequently activated by stress and has been ascribed with either pro- or anti-autophagic functions, depending on the cellular context. Our results demonstrate that activation of the IKK (IkappaB kinase) complex, which is critical for the stress-elicited activation of NFkappaB, is sufficient to promote autophagy independent of NFkappaB, and that IKK is required for the optimal induction of autophagy by both physiological and pharmacological autophagic triggers.


Subject(s)
Autophagy/physiology , I-kappa B Kinase/physiology , Stress, Physiological/physiology , Animals , Autophagy/genetics , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Models, Biological , Signal Transduction/genetics , Signal Transduction/physiology , Stress, Physiological/genetics
16.
EMBO J ; 29(3): 619-31, 2010 Feb 03.
Article in English | MEDLINE | ID: mdl-19959994

ABSTRACT

In response to stress, cells start transcriptional and transcription-independent programs that can lead to adaptation or death. Here, we show that multiple inducers of autophagy, including nutrient depletion, trigger the activation of the IKK (IkappaB kinase) complex that is best known for its essential role in the activation of the transcription factor NF-kappaB by stress. Constitutively active IKK subunits stimulated autophagy and transduced multiple signals that operate in starvation-induced autophagy, including the phosphorylation of AMPK and JNK1. Genetic inhibition of the nuclear translocation of NF-kappaB or ablation of the p65/RelA NF-kappaB subunit failed to suppress IKK-induced autophagy, indicating that IKK can promote the autophagic pathway in an NF-kappaB-independent manner. In murine and human cells, knockout and/or knockdown of IKK subunits (but not that of p65) prevented the induction of autophagy in response to multiple stimuli. Moreover, the knockout of IKK-beta suppressed the activation of autophagy by food deprivation or rapamycin injections in vivo, in mice. Altogether, these results indicate that IKK has a cardinal role in the stimulation of autophagy by physiological and pharmacological stimuli.


Subject(s)
Autophagy/physiology , I-kappa B Kinase/physiology , Animals , Autophagy/genetics , Cells, Cultured , HeLa Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Multiprotein Complexes/physiology , NF-kappa B/genetics , NF-kappa B/metabolism , NIH 3T3 Cells , Signal Transduction/physiology
17.
Apoptosis ; 13(9): 1148-61, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18670883

ABSTRACT

High-risk myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) are characterized by the activation of the anti-apoptotic transcription factor NFkappaB, via the IKK complex. Here, we show that constitutive activation of the receptor tyrosine kinase Flt3 is responsible for IKK activation. Chemical inhibition or knockdown of Flt3 with small interfering RNAs reduced NFkappaB activation in MDS and AML cell lines, as well as in primary CD34(+) bone marrow cells from patients, causing apoptosis. Epistatic analysis involving the simultaneous inhibition of Flt3 and IKK suggested that both kinases act in the same anti-apoptotic pathway. An IKK2 mutant with a constitutive kinase activity and a plasma membrane-tethered mutant of NEMO that activates IKK1/2 prevented the cytocidal action of Flt3 inhibition. Flt3 phosphorylates IKK2 in vitro, and Flt3 inhibition reduced the phosphorylation of IKK2 in MDS or AML cell lines. IKK2 and Flt3 physically associated in MDS and AML cells, and Flt3 inhibition disrupted this interaction. Flt3 inhibition only killed CD34(+) bone marrow cells from high-risk MDS and AML patients, in correlation with blast numbers and NFkappaB activity, yet had no lethal effect on healthy CD34(+) cells or cells from low-risk MDS. These results suggest that Flt3 inhibitors might exert an anti-neoplastic effect in high-risk MDS and AML through inhibition of NFkappaB.


Subject(s)
Leukemia, Myeloid, Acute/enzymology , Myelodysplastic Syndromes/enzymology , NF-kappa B/metabolism , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Apoptosis , Bone Marrow Cells/enzymology , Bone Marrow Cells/pathology , Cell Line, Tumor , Epistasis, Genetic , Humans , I-kappa B Kinase/metabolism , Middle Aged
18.
Cell Cycle ; 7(14): 2139-45, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18641459

ABSTRACT

Malignant myeloblasts arising in high-risk myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) are characterized by the constitutive activation of the anti-apoptotic transcription factor NFkappaB. We found that DNA methyltransferase (DNMT) inhibitors (such as azacytidine and 5-aza-2'-deoxycytidine) and histone deacetylase (HDAC) inhibitors (such as trichostatin and valproic acid) efficiently induced apoptosis in the P39 MDS/AML cell line, correlating with an inhibition of NFkappaB (which translocated from the nucleus to the cytoplasm). This effect was obtained rapidly, within a few hours, suggesting that it was not due to epigenetic reprogramming. Indeed, DNMT and HDAC inhibitors reduced the phosphorylation of the NFkappaB-activating kinase IKKalpha/beta, and this effect was also observed in enucleated cells. Finally, circulating myeloblasts from AML patients treated with the DNMT inhibitor 5-aza-2'-deoxycytidine manifested a rapid (2 hours post-treatment) inhibition of NFkappaB and IKKalpha/beta. Altogether, these results indicate that DNMT and HDAC inhibitors can inhibit the constitutive activation of NFkappaB in malignant myeloblasts in vitro and in vivo through a novel mechanism.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Granulocyte Precursor Cells/drug effects , Granulocyte Precursor Cells/metabolism , Histone Deacetylase Inhibitors , NF-kappa B/antagonists & inhibitors , Azacitidine/pharmacology , Cell Line, Tumor , Female , Granulocyte Precursor Cells/enzymology , Humans , Hydroxamic Acids/pharmacology , I-kappa B Kinase/metabolism , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Valproic Acid/pharmacology
19.
Biochem Pharmacol ; 76(11): 1417-25, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-18617157

ABSTRACT

Erlotinib and gefitinib, two inhibitors of the epidermal growth factor receptor (EGFR), can stimulate apoptosis and differentiation of myeloid cell lines that lack EGFR, unveiling a novel, therapeutically exploitable off-target effect of tyrosine kinase inhibitors. Here, we performed a side-by-side comparison of erlotinib and gefitinib effects on a broad spectrum of malignant myeloid cell lines, as well as on primary myeloblasts freshly purified from the bone marrow of patients with myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). Both erlotinib and gefitinib induce apoptosis of a cell line (KG-1) that represents AML, and differentiation in another cell line (P39) derived from a patient with high-risk MDS. In this setting, erlotinib was more efficient than gefitinib. Erlotinib and gefitinib were equipotent in inducing apoptosis of primary CD34+ myeloblasts from MDS and AML patients, yet had no toxic effect on CD34+ progenitor cells from healthy donors. Although the response of individual MDS and AML patients in vitro was highly heterogeneous, the pro-apoptotic effects of erlotinib and gefitinib correlated significantly. These results suggest that erlotinib and gefitinib share a mechanistically related off-target effect that may be taken advantage of for the therapy of MDS and AML.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Adult , Aged , Aged, 80 and over , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Erlotinib Hydrochloride , Female , Gefitinib , Humans , Male , Middle Aged , Quinazolines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...