Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Exp Med ; 221(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38385946

ABSTRACT

Cellular senescence is a critical stress response program implicated in embryonic development, wound healing, aging, and immunity, and it backs up apoptosis as an ultimate cell-cycle exit mechanism. In analogy to replicative exhaustion of telomere-eroded cells, premature types of senescence-referring to oncogene-, therapy-, or virus-induced senescence-are widely considered irreversible growth arrest states as well. We discuss here that entry into full-featured senescence is not necessarily a permanent endpoint, but dependent on essential maintenance components, potentially transient. Unlike a binary state switch, we view senescence with its extensive epigenomic reorganization, profound cytomorphological remodeling, and distinctive metabolic rewiring rather as a journey toward a full-featured arrest condition of variable strength and depth. Senescence-underlying maintenance-essential molecular mechanisms may allow cell-cycle reentry if not continuously provided. Importantly, senescent cells that resumed proliferation fundamentally differ from those that never entered senescence, and hence would not reflect a reversion but a dynamic progression to a post-senescent state that comes with distinct functional and clinically relevant ramifications.


Subject(s)
Aging , Cellular Senescence , Female , Pregnancy , Humans , Cell Cycle , Cell Division , Apoptosis
2.
Nature ; 599(7884): 283-289, 2021 11.
Article in English | MEDLINE | ID: mdl-34517409

ABSTRACT

Derailed cytokine and immune cell networks account for the organ damage and the clinical severity of COVID-19 (refs. 1-4). Here we show that SARS-CoV-2, like other viruses, evokes cellular senescence as a primary stress response in infected cells. Virus-induced senescence (VIS) is indistinguishable from other forms of cellular senescence and is accompanied by a senescence-associated secretory phenotype (SASP), which comprises pro-inflammatory cytokines, extracellular-matrix-active factors and pro-coagulatory mediators5-7. Patients with COVID-19 displayed markers of senescence in their airway mucosa in situ and increased serum levels of SASP factors. In vitro assays demonstrated macrophage activation with SASP-reminiscent secretion, complement lysis and SASP-amplifying secondary senescence of endothelial cells, which mirrored hallmark features of COVID-19 such as macrophage and neutrophil infiltration, endothelial damage and widespread thrombosis in affected lung tissue1,8,9. Moreover, supernatant from VIS cells, including SARS-CoV-2-induced senescence, induced neutrophil extracellular trap formation and activation of platelets and the clotting cascade. Senolytics such as navitoclax and a combination of dasatinib plus quercetin selectively eliminated VIS cells, mitigated COVID-19-reminiscent lung disease and reduced inflammation in SARS-CoV-2-infected hamsters and mice. Our findings mark VIS as a pathogenic trigger of COVID-19-related cytokine escalation and organ damage, and suggest that senolytic targeting of virus-infected cells is a treatment option against SARS-CoV-2 and perhaps other viral infections.


Subject(s)
COVID-19 Drug Treatment , COVID-19/pathology , COVID-19/virology , Cellular Senescence/drug effects , Molecular Targeted Therapy , SARS-CoV-2/pathogenicity , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Animals , COVID-19/complications , Cell Line , Cricetinae , Dasatinib/pharmacology , Dasatinib/therapeutic use , Disease Models, Animal , Female , Humans , Male , Mice , Quercetin/pharmacology , Quercetin/therapeutic use , SARS-CoV-2/drug effects , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Thrombosis/complications , Thrombosis/immunology , Thrombosis/metabolism
3.
Blood Cancer Discov ; 2(1): 70-91, 2021 01.
Article in English | MEDLINE | ID: mdl-33447829

ABSTRACT

Based on gene expression profiles, diffuse large B cell lymphoma (DLBCL) is sub-divided into germinal center B cell-like (GCB) and activated B cell-like (ABC) DLBCL. Two of the most common genomic aberrations in ABC-DLBCL are mutations in MYD88, as well as BCL2 copy number gains. Here, we employ immune phenotyping, RNA-Seq and whole exome sequencing to characterize a Myd88 and Bcl2-driven mouse model of ABC-DLBCL. We show that this model resembles features of human ABC-DLBCL. We further demonstrate an actionable dependence of our murine ABC-DLBCL model on BCL2. This BCL2 dependence was also detectable in human ABC-DLBCL cell lines. Moreover, human ABC-DLBCLs displayed increased PD-L1 expression, compared to GCB-DLBCL. In vivo experiments in our ABC-DLBCL model showed that combined venetoclax and RMP1-14 significantly increased the overall survival of lymphoma bearing animals, indicating that this combination may be a viable option for selected human ABC-DLBCL cases harboring MYD88 and BCL2 aberrations.


Subject(s)
Lymphoma, Large B-Cell, Diffuse , Myeloid Differentiation Factor 88 , Animals , Genes, bcl-2 , Germinal Center/metabolism , Lymphoma, Large B-Cell, Diffuse/genetics , Mice , Myeloid Differentiation Factor 88/genetics , Proto-Oncogene Proteins c-bcl-2/genetics
4.
Blood ; 137(20): 2785-2799, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33232972

ABSTRACT

Aberrant B-cell receptor/NF-κB signaling is a hallmark feature of B-cell non-Hodgkin lymphomas, especially in diffuse large B-cell lymphoma (DLBCL). Recurrent mutations in this cascade, for example, in CD79B, CARD11, or NFKBIZ, and also in the Toll-like receptor pathway transducer MyD88, all deregulate NF-κB, but their differential impact on lymphoma development and biology remains to be determined. Here, we functionally investigate primary mouse lymphomas that formed in recipient mice of Eµ-myc transgenic hematopoietic stem cells stably transduced with naturally occurring NF-κB mutants. Although most mutants supported Myc-driven lymphoma formation through repressed apoptosis, CARD11- or MyD88-mutant lymphoma cells selectively presented with a macrophage-activating secretion profile, which, in turn, strongly enforced transforming growth factor ß (TGF-ß)-mediated senescence in the lymphoma cell compartment. However, MyD88- or CARD11-mutant Eµ-myc lymphomas exhibited high-level expression of the immune-checkpoint mediator programmed cell death ligand 1 (PD-L1), thus preventing their efficient clearance by adaptive host immunity. Conversely, these mutant-specific dependencies were therapeutically exploitable by anti-programmed cell death 1 checkpoint blockade, leading to direct T-cell-mediated lysis of predominantly but not exclusively senescent lymphoma cells. Importantly, mouse-based mutant MyD88- and CARD11-derived signatures marked DLBCL subgroups exhibiting mirroring phenotypes with respect to the triad of senescence induction, macrophage attraction, and evasion of cytotoxic T-cell immunity. Complementing genomic subclassification approaches, our functional, cross-species investigation unveils pathogenic principles and therapeutic vulnerabilities applicable to and testable in human DLBCL subsets that may inform future personalized treatment strategies.


Subject(s)
Adaptive Immunity , CARD Signaling Adaptor Proteins/genetics , Cellular Senescence/physiology , Guanylate Cyclase/genetics , Lymphoma, Large B-Cell, Diffuse/immunology , Myeloid Differentiation Factor 88/genetics , Neoplasm Proteins/genetics , T-Lymphocytes, Cytotoxic/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , B7-H1 Antigen/antagonists & inhibitors , CD79 Antigens/genetics , Cell Line, Tumor , Chemotaxis , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genes, Reporter , Genes, myc , Humans , Immune Checkpoint Inhibitors , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/therapy , Macrophages/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense , NF-kappa B/genetics , NF-kappa B/metabolism , Point Mutation , Programmed Cell Death 1 Ligand 2 Protein/antagonists & inhibitors , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Transcriptome
5.
Nat Commun ; 11(1): 3651, 2020 07 20.
Article in English | MEDLINE | ID: mdl-32686676

ABSTRACT

Lesion-based targeting strategies underlie cancer precision medicine. However, biological principles - such as cellular senescence - remain difficult to implement in molecularly informed treatment decisions. Functional analyses in syngeneic mouse models and cross-species validation in patient datasets might uncover clinically relevant genetics of biological response programs. Here, we show that chemotherapy-exposed primary Eµ-myc transgenic lymphomas - with and without defined genetic lesions - recapitulate molecular signatures of patients with diffuse large B-cell lymphoma (DLBCL). Importantly, we interrogate the murine lymphoma capacity to senesce and its epigenetic control via the histone H3 lysine 9 (H3K9)-methyltransferase Suv(ar)39h1 and H3K9me3-active demethylases by loss- and gain-of-function genetics, and an unbiased clinical trial-like approach. A mouse-derived senescence-indicating gene signature, termed "SUVARness", as well as high-level H3K9me3 lymphoma expression, predict favorable DLBCL patient outcome. Our data support the use of functional genetics in transgenic mouse models to incorporate basic biology knowledge into cancer precision medicine in the clinic.


Subject(s)
Cellular Senescence , Histone Methyltransferases , Lymphoma, Large B-Cell, Diffuse , 3T3 Cells , Animals , Cell Line, Tumor , Disease Models, Animal , Epigenesis, Genetic , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Histone Methyltransferases/genetics , Histone Methyltransferases/metabolism , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, Transgenic , Prognosis
7.
Cancer Cell ; 33(2): 322-336.e8, 2018 02 12.
Article in English | MEDLINE | ID: mdl-29438700

ABSTRACT

Oncogene-induced senescence, e.g., in melanocytic nevi, terminates the expansion of pre-malignant cells via transcriptional silencing of proliferation-related genes due to decoration of their promoters with repressive trimethylated histone H3 lysine 9 (H3K9) marks. We show here that structurally distinct H3K9-active demethylases-the lysine-specific demethylase-1 (LSD1) and several Jumonji C domain-containing moieties (such as JMJD2C)-disable senescence and permit Ras/Braf-evoked transformation. In mouse and zebrafish models, enforced LSD1 or JMJD2C expression promoted Braf-V600E-driven melanomagenesis. A large subset of established melanoma cell lines and primary human melanoma samples presented with a collective upregulation of related and unrelated H3K9 demethylase activities, whose targeted inhibition restored senescence, even in Braf inhibitor-resistant melanomas, evoked secondary immune effects and controlled tumor growth in vivo.


Subject(s)
Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Melanoma/genetics , Animals , Histones/metabolism , Humans , Lysine/genetics , Lysine/metabolism , Methylation , Mice, Nude , Promoter Regions, Genetic/genetics
8.
Nature ; 553(7686): 96-100, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29258294

ABSTRACT

Cellular senescence is a stress-responsive cell-cycle arrest program that terminates the further expansion of (pre-)malignant cells. Key signalling components of the senescence machinery, such as p16INK4a, p21CIP1 and p53, as well as trimethylation of lysine 9 at histone H3 (H3K9me3), also operate as critical regulators of stem-cell functions (which are collectively termed 'stemness'). In cancer cells, a gain of stemness may have profound implications for tumour aggressiveness and clinical outcome. Here we investigated whether chemotherapy-induced senescence could change stem-cell-related properties of malignant cells. Gene expression and functional analyses comparing senescent and non-senescent B-cell lymphomas from Eµ-Myc transgenic mice revealed substantial upregulation of an adult tissue stem-cell signature, activated Wnt signalling, and distinct stem-cell markers in senescence. Using genetically switchable models of senescence targeting H3K9me3 or p53 to mimic spontaneous escape from the arrested condition, we found that cells released from senescence re-entered the cell cycle with strongly enhanced and Wnt-dependent clonogenic growth potential compared to virtually identical populations that had been equally exposed to chemotherapy but had never been senescent. In vivo, these previously senescent cells presented with a much higher tumour initiation potential. Notably, the temporary enforcement of senescence in p53-regulatable models of acute lymphoblastic leukaemia and acute myeloid leukaemia was found to reprogram non-stem bulk leukaemia cells into self-renewing, leukaemia-initiating stem cells. Our data, which are further supported by consistent results in human cancer cell lines and primary samples of human haematological malignancies, reveal that senescence-associated stemness is an unexpected, cell-autonomous feature that exerts its detrimental, highly aggressive growth potential upon escape from cell-cycle blockade, and is enriched in relapse tumours. These findings have profound implications for cancer therapy, and provide new mechanistic insights into the plasticity of cancer cells.


Subject(s)
Cellular Reprogramming , Cellular Senescence , Lymphoma, B-Cell/pathology , Neoplastic Stem Cells/pathology , Animals , Biomarkers/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Reprogramming/drug effects , Cellular Senescence/drug effects , Cellular Senescence/genetics , Clone Cells/drug effects , Clone Cells/pathology , Female , Humans , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/genetics , Male , Mice , Mice, Transgenic , Neoplastic Stem Cells/drug effects , Phenotype , Wnt Signaling Pathway/drug effects
9.
Blood ; 129(1): 71-81, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27733358

ABSTRACT

Classical Hodgkin lymphoma (cHL), although originating from B cells, is characterized by the virtual lack of gene products whose expression constitutes the B-cell phenotype. Epigenetic repression of B-cell-specific genes via promoter hypermethylation and histone deacetylation as well as compromised expression of B-cell-committed transcription factors were previously reported to contribute to the lost B-cell phenotype in cHL. Restoring the B-cell phenotype may not only correct a central malignant property, but it may also render cHL susceptible to clinically established antibody therapies targeting B-cell surface receptors or small compounds interfering with B-cell receptor signaling. We conducted a high-throughput pharmacological screening based on >28 000 compounds in cHL cell lines carrying a CD19 reporter to identify drugs that promote reexpression of the B-cell phenotype. Three chemicals were retrieved that robustly enhanced CD19 transcription. Subsequent chromatin immunoprecipitation-based analyses indicated that action of 2 of these compounds was associated with lowered levels of the transcriptionally repressive lysine 9-trimethylated histone H3 mark at the CD19 promoter. Moreover, the antileukemia agents all-trans retinoic acid and arsenic trioxide (ATO) were found to reconstitute the silenced B-cell transcriptional program and reduce viability of cHL cell lines. When applied in combination with a screening-identified chemical, ATO evoked reexpression of the CD20 antigen, which could be further therapeutically exploited by enabling CD20 antibody-mediated apoptosis of cHL cells. Furthermore, restoration of the B-cell phenotype also rendered cHL cells susceptible to the B-cell non-Hodgkin lymphoma-tailored small-compound inhibitors ibrutinib and idelalisib. In essence, we report here a conceptually novel, redifferentiation-based treatment strategy for cHL.


Subject(s)
Antineoplastic Agents/pharmacology , B-Lymphocytes/immunology , Cell Differentiation/drug effects , Hodgkin Disease/immunology , Transcriptome/drug effects , Antigens, CD19/immunology , Antigens, CD20/immunology , B-Lymphocytes/drug effects , Chromatin Immunoprecipitation , Flow Cytometry , High-Throughput Screening Assays , Humans , Phenotype , Polymerase Chain Reaction , Tumor Cells, Cultured
10.
Blood ; 127(22): 2732-41, 2016 06 02.
Article in English | MEDLINE | ID: mdl-27048211

ABSTRACT

The adaptor protein MYD88 is critical for relaying activation of Toll-like receptor signaling to NF-κB activation. MYD88 mutations, particularly the p.L265P mutation, have been described in numerous distinct B-cell malignancies, including diffuse large B-cell lymphoma (DLBCL). Twenty-nine percent of activated B-cell-type DLBCL (ABC-DLBCL), which is characterized by constitutive activation of the NF-κB pathway, carry the p.L265P mutation. In addition, ABC-DLBCL frequently displays focal copy number gains affecting BCL2 Here, we generated a novel mouse model in which Cre-mediated recombination, specifically in B cells, leads to the conditional expression of Myd88(p.L252P) (the orthologous position of the human MYD88(p.L265P) mutation) from the endogenous locus. These mice develop a lymphoproliferative disease and occasional transformation into clonal lymphomas. The clonal disease displays the morphologic and immunophenotypical characteristics of ABC-DLBCL. Lymphomagenesis can be accelerated by crossing in a further novel allele, which mediates conditional overexpression of BCL2 Cross-validation experiments in human DLBCL samples revealed that both MYD88 and CD79B mutations are substantially enriched in ABC-DLBCL compared with germinal center B-cell DLBCL. Furthermore, analyses of human DLBCL genome sequencing data confirmed that BCL2 amplifications frequently co-occurred with MYD88 mutations, further validating our approach. Finally, in silico experiments revealed that MYD88-mutant ABC-DLBCL cells in particular display an actionable addiction to BCL2. Altogether, we generated a novel autochthonous mouse model of ABC-DLBCL that could be used as a preclinical platform for the development and validation of novel therapeutic approaches for the treatment of ABC-DLBCL.


Subject(s)
B-Lymphocytes/metabolism , Cell Transformation, Neoplastic/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Mutation, Missense , Myeloid Differentiation Factor 88/biosynthesis , Neoplasms, Experimental/metabolism , Animals , B-Lymphocytes/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Humans , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Mice, Transgenic , Myeloid Differentiation Factor 88/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics
11.
Nature ; 501(7467): 421-5, 2013 Sep 19.
Article in English | MEDLINE | ID: mdl-23945590

ABSTRACT

Activated oncogenes and anticancer chemotherapy induce cellular senescence, a terminal growth arrest of viable cells characterized by S-phase entry-blocking histone 3 lysine 9 trimethylation (H3K9me3). Although therapy-induced senescence (TIS) improves long-term outcomes, potentially harmful properties of senescent tumour cells make their quantitative elimination a therapeutic priority. Here we use the Eµ-myc transgenic mouse lymphoma model in which TIS depends on the H3K9 histone methyltransferase Suv39h1 to show the mechanism and therapeutic exploitation of senescence-related metabolic reprogramming in vitro and in vivo. After senescence-inducing chemotherapy, TIS-competent lymphomas but not TIS-incompetent Suv39h1(-) lymphomas show increased glucose utilization and much higher ATP production. We demonstrate that this is linked to massive proteotoxic stress, which is a consequence of the senescence-associated secretory phenotype (SASP) described previously. SASP-producing TIS cells exhibited endoplasmic reticulum stress, an unfolded protein response (UPR), and increased ubiquitination, thereby targeting toxic proteins for autophagy in an acutely energy-consuming fashion. Accordingly, TIS lymphomas, unlike senescence models that lack a strong SASP response, were more sensitive to blocking glucose utilization or autophagy, which led to their selective elimination through caspase-12- and caspase-3-mediated endoplasmic-reticulum-related apoptosis. Consequently, pharmacological targeting of these metabolic demands on TIS induction in vivo prompted tumour regression and improved treatment outcomes further. These findings unveil the hypercatabolic nature of TIS that is therapeutically exploitable by synthetic lethal metabolic targeting.


Subject(s)
Autophagy , Cellular Senescence , Glucose/metabolism , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/metabolism , Animals , Apoptosis/drug effects , Autophagy/drug effects , Caspase 12/metabolism , Caspase 3/metabolism , Cellular Senescence/drug effects , Disease Models, Animal , Endoplasmic Reticulum Stress , Female , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Male , Mice , Mice, Transgenic , Proteolysis , Stress, Physiological , Survival Rate
12.
Blood ; 121(15): 2964-74, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23403624

ABSTRACT

Pharmacological strategies capable of directly targeting MYC are elusive. Previous studies have shown that MYC-driven lymphomagenesis is associated with mammalian target of rapamycin (mTOR) activation and a MYC-evoked DNA damage response (DDR) transduced by phosphatidylinositol-3-kinase (PI3K)-related kinases (DNA-PK, ATM, and ATR). Here we report that BEZ235, a multitargeted pan-PI3K/dual-mTOR inhibitor, potently killed primary Myc-driven B-cell lymphomas and human cell lines bearing IG-cMYC translocations. Using pharmacologic and genetic dissection of PI3K/mTOR signaling, dual DDR/mTORC1 inhibition was identified as a key mediator of apoptosis. Moreover, apoptosis was initiated at drug concentrations insufficient to antagonize PI3K/mTORC2-regulated AKT phosphorylation. p53-independent induction of the proapoptotic BH3-only protein BMF was identified as a mechanism by which dual DDR/mTORC1 inhibition caused lymphoma cell death. BEZ235 treatment induced apoptotic tumor regressions in vivo that correlated with suppression of mTORC1-regulated substrates and reduced H2AX phosphorylation and also with feedback phosphorylation of AKT. These mechanistic studies hold important implications for the use of multitargeted PI3K inhibitors in the treatment of hematologic malignancies. In particular, the newly elucidated role of PI3K-related DDR kinases in response to PI3K inhibitors offers a novel therapeutic opportunity for the treatment of hematologic malignancies with an MYC-driven DDR.


Subject(s)
Apoptosis/drug effects , Imidazoles/pharmacology , Lymphoma, B-Cell/prevention & control , Quinolines/pharmacology , Animals , Blotting, Western , Cell Line , DNA Damage/drug effects , Discoidin Domain Receptor 1 , Dose-Response Relationship, Drug , Flow Cytometry , Histones/metabolism , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Survival Analysis , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured
13.
Genes Dev ; 25(20): 2137-46, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21979374

ABSTRACT

In malignancies, enhanced nuclear factor-κB (NF-κB) activity is largely viewed as an oncogenic property that also confers resistance to chemotherapy. Recently, NF-κB has been postulated to participate in a senescence-associated and possibly senescence-reinforcing cytokine response, thereby suggesting a tumor-restraining role for NF-κB. Using a mouse lymphoma model and analyzing transcriptome and clinical data from lymphoma patients, we show here that therapy-induced senescence presents with and depends on active NF-κB signaling, whereas NF-κB simultaneously promotes resistance to apoptosis. Further characterization and genetic engineering of primary mouse lymphomas according to distinct NF-κB-related oncogenic networks reminiscent of diffuse large B-cell lymphoma (DLBCL) subtypes guided us to identify Bcl2-overexpressing germinal center B-cell-like (GCB) DLBCL as a clinically relevant subgroup with significantly superior outcome when NF-κB is hyperactive. Our data illustrate the power of cross-species investigations to functionally test genetic mechanisms in transgenic mouse tumors that recapitulate distinct features of the corresponding human entity, and to ultimately use the mouse model-derived genetic information to redefine novel, clinically relevant patient subcohorts.


Subject(s)
Antineoplastic Agents/pharmacology , Cellular Senescence/drug effects , Cellular Senescence/physiology , NF-kappa B/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Non-Hodgkin/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction
14.
Semin Cancer Biol ; 21(6): 377-84, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22019769

ABSTRACT

Ras/Raf-prototypic oncogenes induce cellular senescence, a terminal cell-cycle arrest, as a default cellular safeguard program, while oncogenic Myc is known to rather promote apoptosis as the prime failsafe mechanism. We review and discuss here evidence for Myc-induced senescence - which is detectable to a limited degree as a cell-autonomous, direct response to Myc action, but occurs predominantly in a non-cell-autonomous fashion via crosstalk of the oncogene-driven cell population with non-neoplastic bystanders, namely cells of the host immune system, prompting them to release pro-senescent cytokines that strike back onto adjacent proliferating tumor cells. In particular, we discuss how Myc-evoked apoptosis serves as a signal for macrophage attraction and activation, followed by the secretion of TGF-ß as a cytokine that is capable of terminally arresting Myc-driven lymphoma cells without causing further DNA damage and without launching a senescence-associated, pro-inflammatory, and, therefore, potentially detrimental cytokine response in the target population. In essence, non-cell-autonomous but still oncogene-orchestrated senescence is a functionally relevant, robustly tumor-suppressive principle with critical implications for conceptually novel anti-cancer therapies in the clinic.


Subject(s)
Cellular Senescence/genetics , Genes, myc , Oncogenes , Humans
15.
J Mol Med (Berl) ; 89(9): 869-75, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21594578

ABSTRACT

Activated oncogenes evoke cellular fail-safe programs such as apoptosis, senescence, or autophagy to protect the organism from the expansion of damaged and potentially harmful cells. Non-cell-autonomous interactions between tumor cells and nonmalignant bystander cells add to cell-autonomous modes of tumor suppression during tumor development and progression. In particular, the role of stroma or host immune cells converting tumor-generated signals into a response that feeds back to the tumor cell population has been experimentally underappreciated. Using the Eµ-myc transgenic mouse lymphoma model, we elucidated how constitutive Myc signaling indirectly promotes cellular senescence via cytokines that were released by nonmalignant cells in response to oncogene-evoked cell-autonomous effects. Specifically, Myc primarily promotes apoptosis in a subset of the tumor cell population, leading to the attraction of macrophages, which subsequently engulf the apoptotic remainders. Phagocytosis-activated macrophages, in turn, exhibit strongly increased secretion of various cytokines, among them transforming growth factor beta to an extent that is capable of inducing cellular senescence in surrounding malignant cells. Our findings, recapitulated in human aggressive B-cell lymphomas, unveil that apoptosis and senescence are not simply two context-dependent cell-autonomous choices of stress responses, but rather cooperate via extracellular mediators-namely cells of the innate immune system-to profoundly limit tumorigenesis in vivo. A deeper mechanistic understanding of the organismic interconnection between different fail-safe programs will help to identify cellular components of the tumor stroma and their signal mediators that are readily available to impose a second line of host defense against cancer cells. This will open new perspectives for the development of antineoplastic therapies, whose targets not only encompass tumor but also stroma cell populations.


Subject(s)
Apoptosis/genetics , Cellular Senescence/genetics , Neoplasms/genetics , Neoplasms/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Animals , Humans , Immune System/immunology , Immune System/metabolism , Macrophages/immunology , Macrophages/metabolism , Neoplasms/immunology , Stromal Cells/metabolism , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism , Tumor Microenvironment
16.
Cancer Cell ; 17(3): 262-72, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20227040

ABSTRACT

Activated RAS/BRAF oncogenes induce cellular senescence as a tumor-suppressive barrier in early cancer development, at least in part, via an oncogene-evoked DNA damage response (DDR). In contrast, Myc activation-although producing a DDR as well-is known to primarily elicit an apoptotic countermeasure. Using the Emu-myc transgenic mouse lymphoma model, we show here in vivo that apoptotic lymphoma cells activate macrophages to secrete transforming growth factor beta (TGF-beta) as a critical non-cell-autonomous inducer of cellular senescence. Accordingly, neutralization of TGF-beta action, like genetic inactivation of the senescence-related histone methyltransferase Suv39h1, significantly accelerates Myc-driven tumor development via cancellation of cellular senescence. These findings, recapitulated in human aggressive B cell lymphomas, demonstrate that tumor-prompted stroma-derived signals may limit tumorigenesis by feedback senescence induction.


Subject(s)
Cellular Senescence/physiology , Lymphoma/metabolism , Methyltransferases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Repressor Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Apoptosis , Lymphoma/pathology , Macrophage Activation , Macrophages/metabolism , Methyltransferases/physiology , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-myc/physiology , Repressor Proteins/physiology , Tumor Suppressor Protein p53/physiology
17.
Blood ; 110(8): 2996-3004, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17562874

ABSTRACT

In addition to the ARF/p53 pathway, the DNA damage response (DDR) has been recognized as another oncogene-provoked anticancer barrier in early human tumorigenesis leading to apoptosis or cellular senescence. DDR mutations may promote tumor formation, but their impact on treatment outcome remains unclear. In this study, we generated ataxia telangiectasia mutated (Atm)-proficient and -deficient B-cell lymphomas in Emu-myc transgenic mice to examine the role of DDR defects in lymphomagenesis and treatment sensitivity. Atm inactivation accelerated development of lymphomas, and their DNA damage checkpoint defects were virtually indistinguishable from those observed in Atm+/+-derived lymphomas that spontaneously inactivated the proapoptotic Atm/p53 cascade in response to Myc-evoked reactive oxygen species (ROS). Importantly, acquisition of DDR defects, but not selection against the ARF pathway, could be prevented by lifelong exposure to the ROS scavenger N-acetylcysteine (NAC) in vivo. Following anticancer therapy, DDR-compromised lymphomas displayed apoptotic but, surprisingly, no senescence defects and achieved a much poorer long-term outcome when compared with DDR-competent lymphomas treated in vivo. Hence, Atm eliminates preneoplastic lesions by converting oncogenic signaling into apoptosis, and selection against an Atm-dependent response promotes formation of lymphomas with predetermined treatment insensitivity.


Subject(s)
Cell Cycle Proteins/genetics , DNA Damage , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/genetics , Genes, myc , Lymphoma, B-Cell/genetics , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/genetics , Animals , Apoptosis/physiology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA-Binding Proteins/metabolism , Flow Cytometry , Gene Transfer Techniques , Immunoblotting , Lymphoma, B-Cell/drug therapy , Mice , Mice, Transgenic , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering , Reactive Oxygen Species/pharmacology , Transduction, Genetic , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
18.
J Biol Chem ; 278(35): 32587-95, 2003 Aug 29.
Article in English | MEDLINE | ID: mdl-12813031

ABSTRACT

The concept that the tumor suppressor p53 is a latent DNA-binding protein that must become activated for sequence-specific DNA binding recently has been challenged, although the "activation" phenomenon has been well established in in vitro DNA binding assays. Using electrophoretic mobility shift assays and fluorescence correlation spectroscopy, we analyzed the binding of "latent" and "activated" p53 to double-stranded DNA oligonucleotides containing or not containing a p53 consensus binding site (DNAspec or DNAunspec, respectively). In the absence of competitor DNA, latent p53 bound DNAspec and DNAunspec with high affinity in a sequence-independent manner. Activation of p53 by the addition of the C-terminal antibody PAb421 significantly decreased the binding affinity for DNAunspec and concomitantly increased the binding affinity for DNAspec. The net result of this dual effect is a significant difference in the affinity of activated p53 for DNAspec and DNAunspec, which explains the activation of p53. High affinity nonspecific DNA binding of latent p53 required both the p53 core domain and the p53 C terminus, whereas high affinity sequence-specific DNA binding of activated p53 was mediated by the p53 core domain alone. The data suggest that high affinity nonspecific DNA binding of latent and high affinity sequence-specific binding of activated p53 to double-stranded DNA differ in their requirement for the C terminus and involve different structural features of the core domain. Because high affinity nonspecific DNA binding of latent p53 is restricted to wild type p53, we propose that it relates to its tumor suppressor functions.


Subject(s)
Spectrometry, Fluorescence/methods , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism , Animals , Antibodies, Monoclonal/metabolism , DNA/metabolism , Dose-Response Relationship, Drug , Epitopes , Humans , Kinetics , Mice , Microscopy, Fluorescence , Mutation , Oligonucleotides/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry
19.
J Biol Chem ; 277(43): 41192-203, 2002 Oct 25.
Article in English | MEDLINE | ID: mdl-12171916

ABSTRACT

Transcriptional activation of p53-regulated genes is initiated by sequence-specific DNA binding of p53 to target binding sites. Regulation of sequence-specific DNA binding is complex and occurs at various levels. We demonstrate that DNA topology is an important parameter for regulating the selective and highly specific interaction of p53 with its target binding sites. Specific binding of wild-type p53 is greatly enhanced when cognate binding sites are present in a non-linear stem-loop conformation. The C-terminal domain plays a key role in regulating the specific interactions of p53 with target binding sites in a DNA conformation-dependent manner. The C-terminal domain is required for binding to target sites in a non-linear DNA conformation in contrast to the strong inhibitory effects of the C terminus on p53 interaction with linear DNA. We propose that selective binding of p53 to various promoters may be determined by the DNA conformation within p53 cognate sites.


Subject(s)
DNA/metabolism , Nucleic Acid Conformation , Tumor Suppressor Protein p53/metabolism , Base Sequence , Binding Sites , DNA/chemistry , DNA Primers , Electrophoretic Mobility Shift Assay , Molecular Sequence Data , Recombinant Proteins/metabolism , Tumor Suppressor Protein p53/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...