Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
J Exp Clin Cancer Res ; 42(1): 203, 2023 Aug 10.
Article in English | MEDLINE | ID: mdl-37563605

ABSTRACT

BACKGROUND: TP53, encoding the tumor suppressor p53, is frequently mutated in various cancers, producing mutant p53 proteins (mutp53) which can exhibit neomorphic, gain-of-function properties. The latter transform p53 into an oncoprotein that promotes metastatic tumor progression via downstream effectors such as ENTPD5, an endoplasmic reticulum UDPase involved in the calnexin/calreticulin cycle of N-glycoprotein biosynthesis. Elucidating the mechanisms underlying the pro-metastatic functions of the mutp53-ENTPD5 axis is crucial for developing targeted therapies for aggressive metastatic cancer. METHODS: We analyzed pancreatic, lung, and breast adenocarcinoma cells with p53 missense mutations to study the impact of mutp53 and ENTPD5 on the N-glycoproteins integrin-α5 (ITGA5) and integrin-ß1 (ITGB1), which heterodimerize to form the key fibronectin receptor. We assessed the role of the mutp53-ENTPD5 axis in integrin-dependent tumor-stroma interactions and tumor cell motility using adhesion, migration, and invasion assays, identifying and validating therapeutic intervention targets. We employed an orthotopic xenograft model of pancreatic ductal adenocarcinoma to examine in vivo targeting of mutp53-ENTPD5-mediated ITGA5 regulation for cancer therapy. RESULTS: Mutp53 depletion diminished ITGA5 and ITGB1 expression and impaired tumor cell adhesion, migration, and invasion, rescued by ENTPD5. The mutp53-ENTPD5 axis maintained ITGA5 expression and function via the calnexin/calreticulin cycle. Targeting this axis using ITGA5-blocking antibodies, α-glucosidase inhibitors, or pharmacological degradation of mutp53 by HSP90 inhibitors, such as Ganetespib, effectively inhibited ITGA5-mediated cancer cell motility in vitro. In the orthotopic xenograft model, Ganetespib reduced ITGA5 expression and metastasis in an ENTPD5-dependent manner. CONCLUSIONS: The mutp53-ENTPD5 axis fosters ITGA5 and ITGB1 expression and tumor cell motility through the calnexin/calreticulin cycle, contributing to cancer metastasis. ITGA5-blocking antibodies or α-glucosidase inhibitors target this axis and represent potential therapeutic options worth exploring in preclinical models. The pharmacologic degradation of mutp53 by HSP90 inhibitors effectively blocks ENTPD5-ITGA5-mediated cancer cell motility and metastasis in vivo, warranting further clinical evaluation in p53-mutant cancers. This research underscores the significance of understanding the complex interplay between mutp53, ENTPD5, and the calnexin/calreticulin cycle in integrin-mediated metastatic tumor progression, offering valuable insights for the development of potential therapeutic strategies.


Subject(s)
Adenocarcinoma , Antineoplastic Agents , Animals , Humans , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Calnexin/genetics , Calnexin/metabolism , Integrin alpha5/metabolism , Calreticulin/metabolism , Antibodies, Blocking/metabolism , Glycoside Hydrolase Inhibitors , Cell Line, Tumor , Molecular Chaperones/metabolism , Disease Models, Animal , Pyrophosphatases/metabolism , Oncogene Proteins/metabolism
2.
J Orthop Traumatol ; 24(1): 6, 2023 Feb 10.
Article in English | MEDLINE | ID: mdl-36765020

ABSTRACT

BACKGROUND: Allograft bone screws are rarely described for the fixation of the scaphoid. When fresh fractures are treated, metal screws are mainly used; when pseudarthrosis is the indication, plates in combination with vascularized or non-vascularized bone grafts are mainly used. The necessity of metallic screw removal is under debate, but it is mandatory for plates because of movement restrictions due to the plate. The use of biomaterials in scaphoid fracture fixation was described as leading to union rates of between 64 and 100%. Brcic showed the incorporation of an allogeneic cortical bone screw at 10 weeks postoperative, along with revascularization and stable osteosynthesis with primary bone healing, without any signs of immunological rejection. The purpose of this retrospective study was to explore the results obtained using an allogenic cortical bone screw (Shark Screw®) in patients with fresh scaphoid fracture fixation and pseudarthroses with respect to union rates and time to union. PATIENTS AND METHODS: We retrospectively analyzed 75 patients: 31 with fresh fractures and 44 pseudarthrosis patients. The Shark Screw® was used for the fixation of the scaphoid in the fresh-fracture and pseudarthrosis patients. We evaluated the union rate, complication rate and time to union. RESULTS: Using the human allogeneic cortical bone screw for scaphoid fracture fixation led to a high union rate (94-96%). There were two nonunions in the fresh fracture group and two nonunions in the pseudarthrosis group. The complication rate was 1.3% (1 patient). Median time to union was 16, 18 and 29 weeks for the fresh-fracture, pseudarthrosis and delayed-union patients, respectively. The treatment of fresh scaphoid fractures and pseudarthroses showed similar union rates to those described in the literature, uses a shorter and less invasive surgical method with no need for hardware removal, and has a low complication rate. CONCLUSION: Using the human allogenic cortical bone screw (Shark Screw®) led to similar union rates in fresh fractures-but better union rates in pseudarthrosis patients-compared to those presented in the literature for other scaphoid fracture fixation techniques, and it enabled a short and low-invasive procedure without any donor site morbidity and without the necessity to remove the hardware in a second surgery. The pseudarthrosis patient group showed a particularly strong benefit from this new procedure. The physiological bone metabolism remodels the cortical bone screw without scars. LEVEL OF EVIDENCE: III: retrospective cohort study, therapeutic investigation of a treatment.


Subject(s)
Fractures, Bone , Fractures, Ununited , Hematopoietic Stem Cell Transplantation , Pseudarthrosis , Scaphoid Bone , Wrist Injuries , Humans , Fractures, Bone/surgery , Pseudarthrosis/surgery , Retrospective Studies , Scaphoid Bone/surgery , Fracture Healing/physiology , Fracture Fixation, Internal/methods , Bone Screws , Cortical Bone
3.
Mol Cancer ; 21(1): 191, 2022 10 03.
Article in English | MEDLINE | ID: mdl-36192757

ABSTRACT

BACKGROUND: In vivo gene editing of somatic cells with CRISPR nucleases has facilitated the generation of autochthonous mouse tumors, which are initiated by genetic alterations relevant to the human disease and progress along a natural timeline as in patients. However, the long and variable, orthotopic tumor growth in inner organs requires sophisticated, time-consuming and resource-intensive imaging for longitudinal disease monitoring and impedes the use of autochthonous tumor models for preclinical studies. METHODS: To facilitate a more widespread use, we have generated a reporter mouse that expresses a Cre-inducible luciferase from Gaussia princeps (GLuc), which is secreted by cells in an energy-consuming process and can be measured quantitatively in the blood as a marker for the viable tumor load. In addition, we have developed a flexible, complementary toolkit to rapidly assemble recombinant adenoviruses (AVs) for delivering Cre recombinase together with CRISPR nucleases targeting cancer driver genes. RESULTS: We demonstrate that intratracheal infection of GLuc reporter mice with CRISPR-AVs efficiently induces lung tumors driven by mutations in the targeted cancer genes and simultaneously activates the GLuc transgene, resulting in GLuc secretion into the blood by the growing tumor. GLuc blood levels are easily and robustly quantified in small-volume blood samples with inexpensive equipment, enable tumor detection already several months before the humane study endpoint and precisely mirror the kinetics of tumor development specified by the inducing gene combination. CONCLUSIONS: Our study establishes blood-based GLuc monitoring as an inexpensive, rapid, high-throughput and animal-friendly method to longitudinally monitor autochthonous tumor growth in preclinical studies.


Subject(s)
Copepoda , Lung Neoplasms , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , Copepoda/genetics , Copepoda/metabolism , Gene Editing , Genes, Reporter , Humans , Luciferases/genetics , Luciferases/metabolism , Lung Neoplasms/genetics , Mice
4.
BMC Cancer ; 22(1): 785, 2022 Jul 18.
Article in English | MEDLINE | ID: mdl-35850701

ABSTRACT

BACKGROUND: More than twenty years after its discovery, the role of the importin beta superfamily member Ran GTP-binding protein (RanBP) 17 is still ill defined. Previously, we observed notable RanBP17 RNA expression levels in head and neck squamous cell carcinoma (HNSCC) cell lines with disruptive TP53 mutations. METHODS: We deployed HNSCC cell lines as well as cell lines from other tumor entities such as HCT116, MDA-MB-231 and H460, which were derived from colon, breast and lung cancers respectively. RNAi was used to evaluate the effect of RanBP17 on cell proliferation. FACS analysis was used for cell sorting according to their respective cell cycle phase and for BrdU assays. Immunocytochemistry was deployed for colocalization studies of RanBP17 with Nucleolin and SC35 (nuclear speckles) domains. TCGA analysis was performed for prognostic assessment and correlation analysis of RanBP17 in HNSCC patients. RESULTS: RNAi knockdown of RanBP17, significantly reduced cell proliferation in HNSCC cell lines. This effect was also seen in the HNSCC unrelated cell lines HCT116 and MDA-MB-231. Similarly, inhibiting cell proliferation with cisplatin reduced RanBP17 in keratinocytes but lead to induction in tumor cell lines. A similar observation was made in tumor cell lines after treatment with the EGFR kinase inhibitor AG1478. In addition to previous reports, showing colocalization of RanBP17 with SC35 domains, we observed colocalization of RanBP17 to nuclear bodies that are distinct from nucleoli and SC35 domains. Interestingly, for HPV positive but not HPV negative HNSCC, TCGA data base analysis revealed a strong positive correlation of RanBP17 RNA with patient survival and CDKN2A. CONCLUSIONS: Our data point to a role of RanBP17 in proliferation of HNSCC and other epithelial cells. Furthermore, RanBP17 could potentially serve as a novel prognostic marker for HNSCC patients. However, we noted a major discrepancy between RanBP17 RNA and protein expression levels with the used antibodies. These observations could be explained by the presence of additional RanBP17 splice isoforms and more so of non-coding circular RanBP17 RNA species. These aspects need to be addressed in more detail by future studies.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation , Head and Neck Neoplasms/genetics , Humans , Protein Kinase Inhibitors/pharmacology , RNA , Squamous Cell Carcinoma of Head and Neck/genetics , beta Karyopherins/genetics , ran GTP-Binding Protein/genetics , ran GTP-Binding Protein/metabolism , ran GTP-Binding Protein/pharmacology
5.
Cells ; 11(11)2022 05 27.
Article in English | MEDLINE | ID: mdl-35681458

ABSTRACT

Autophagy allows cells to temporarily tolerate energy stress by replenishing critical metabolites through self-digestion, thereby attenuating the cytotoxic effects of anticancer drugs that target tumor metabolism. Autophagy defects could therefore mark a metabolically vulnerable cancer state and open a therapeutic window. While mutations of autophagy genes (ATGs) are notably rare in cancer, haploinsufficiency network analyses across many cancers have shown that the autophagy pathway is frequently hit by somatic copy number losses of ATGs such as MAP1LC3B/ATG8F (LC3), BECN1/ATG6 (Beclin-1), and ATG10. Here, we used CRISPR/Cas9 technology to delete increasing numbers of copies of one or more of these ATGs in non-small cell lung cancer cells and examined the effects on sensitivity to compounds targeting aerobic glycolysis, a hallmark of cancer metabolism. Whereas the complete knockout of one ATG blocked autophagy and led to profound metabolic vulnerability, this was not the case for combinations of different nonhomozygous deletions. In cancer patients, the effect of ATG copy number loss was blunted at the protein level and did not lead to the accumulation of p62 as a sign of reduced autophagic flux. Thus, the autophagy pathway is shown to be markedly robust and resilient, even with the concomitant copy number loss of key autophagy genes.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Autophagy/genetics , Beclin-1/genetics , Carcinoma, Non-Small-Cell Lung/genetics , DNA Copy Number Variations/genetics , Humans , Lung Neoplasms/genetics
6.
Cell Death Differ ; 29(11): 2163-2176, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35459909

ABSTRACT

The processes leading from disturbed B-cell development to adult B-cell progenitor acute lymphoblastic leukemia (BCP-ALL) remain poorly understood. Here, we describe Irf4-/- mice as prone to developing BCP-ALL with age. Irf4-/- preB-I cells exhibited impaired differentiation but enhanced proliferation in response to IL-7, along with reduced retention in the IL-7 providing bone marrow niche due to decreased CXCL12 responsiveness. Thus selected, preB-I cells acquired Jak3 mutations, probably following irregular AID activity, resulting in malignant transformation. We demonstrate heightened IL-7 sensitivity due to Jak3 mutants, devise a model to explain it, and describe structural and functional similarities to Jak2 mutations often occurring in human Ph-like ALL. Finally, targeting JAK signaling with Ruxolitinib in vivo prolonged survival of mice bearing established Irf4-/- leukemia. Intriguingly, organ infiltration including leukemic meningeosis was selectively reduced without affecting blood blast counts. In this work, we present spontaneous leukemogenesis following IRF4 deficiency with potential implications for high-risk BCP-ALL in adult humans.


Subject(s)
Burkitt Lymphoma , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Adult , Animals , Humans , Mice , B-Lymphocytes , Burkitt Lymphoma/pathology , Interleukin-7/genetics , Janus Kinase 3/genetics , Mutation/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Signal Transduction
7.
Arch Orthop Trauma Surg ; 141(7): 1207-1213, 2021 Jul.
Article in English | MEDLINE | ID: mdl-32852595

ABSTRACT

PURPOSE: Electric scooters (e-scooters) are an emerging way of mobility in cities around the world. Despite quickly rising numbers of e-scooters, limited studies report on incidence and severity of e-scooter-associated injuries. The aim of our study was to report on these injuries and identify potential protective measures to ultimately decrease e-scooter-associated morbidity. METHODS: We performed a retrospective multicentre study including all patients, who were admitted to three major trauma departments in Vienna from May 2018 to September 2019. We analysed patients' data, including demographics, injury pattern, types of injury and subsequent treatment. RESULTS: A total number of 175 patients (115 males, 60 females) sustained e-scooter-associated injuries. Patients' mean age was 34.4 years [4-74]. While the mean Injury Severity Score (ISS) was 3.4, 11 patients presented with an ISS ≥ 9 and 2 patients with an ISS ≥ 16. ISS increased with age. Older patients (≥ 40 years) presented a significantly higher ISS than younger patients (< 40 years) (P = 0.011). Seventy-one patients (40.6%) sustained major injuries affecting head (35.2%) and upper extremities (36.6%). Twenty-three patients (13.1%) required surgery leading to hospitalization of 11 days on average [1-115]. E-scooter-associated injuries increased during late afternoon plateauing at 8.00 pm. However, the largest share of patients (39.2%) sustained their injuries during early night (8.00 pm to 1.59 am) with especially young adults (19-39 years) being at risk. CONCLUSION: The popularity of rideshare e-scooters across cities worldwide seems to be on the rise, so are e-scooter-associated injuries. These injuries should be considered high-energy trauma affecting primarily head and upper extremity; indeed, 17.7% sustained major head injuries. Therefore, the mandatory use of a helmet seems to be adequate to decrease head injury-associated morbidity. Ultimately, given the remarkably high rates of nighttime injuries, an e-scooter ban during night could further cut injury numbers in half.


Subject(s)
Accidents, Traffic/statistics & numerical data , Wounds and Injuries/epidemiology , Adolescent , Adult , Aged , Austria/epidemiology , Child , Child, Preschool , Female , Head Protective Devices , Humans , Incidence , Injury Severity Score , Male , Middle Aged , Motor Vehicles , Retrospective Studies , Young Adult
8.
Neuroendocrinology ; 111(10): 965-985, 2021.
Article in English | MEDLINE | ID: mdl-33108790

ABSTRACT

INTRODUCTION: Autophagic cell death in cancer cells can be mediated by inhibition of deacetylases. Although extensive studies have focused on the autophagic process in cancer, little is known about the role of autophagy in degrading cytosolic and nuclear components of pancreatic neuroendocrine neoplastic (pNEN) cells leading to cell death, thus improving the therapy of patients affected by pNEN. METHODS: 2D and 3D human pNEN and pancreatic stellate cells were treated with panobinostat and bafilomycin. Autophagy markers were detected by RT-qPCR, immunofluorescence, and Western blot. Autophagosomes were detected by electron microscopy and their maturation by real-time fluorescence of LC3B stable transfected cells. ChIP was performed at the cAMP responsive element. Immunofluorescence was performed in murine pancreatic tissue. RESULTS: We observed that pan-deacetylase inhibitor panobinostat treatment causes autophagic cell death in pNEN cells. We also found that although AMPK-α phosphorylation is counterbalanced by phosphorylated AKT, it is not capable to inhibiting autophagic cell death. However, the binding activity of the cAMP responsive element is prompted by panobinostat. Although autophagy inhibition prevented autophagosome synthesis, maturation, and cell death, panobinostat treatment induced the accumulation of mature autophagosomes in the cytosol and the nucleus, leading to disruption of the organelles, cellular digestion, and decay. Observation of autophagosome membrane proteins Beclin1 and LC3B aggregation in murine pancreatic islets indicates that autophagy restoration may also lead to autophagosome aggregation in murine insulinoma cells. A basal low expression of autophagy markers was detectable in patients affected by pNEN, and, interestingly, the expression of these markers was significantly lower in metastatic pNEN. DISCUSSION/CONCLUSION: Our study highlights that the autophagy functional restoration and prolongation of this catabolic process, mediated by inhibition of deacetylase, is responsible for the reduction of pNEN cells. Prompting of autophagy cell death could be a promising strategy for the therapy of pNEN.


Subject(s)
Autophagic Cell Death/drug effects , Enzyme Inhibitors/pharmacology , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Cell Line, Tumor , Humans , Panobinostat/pharmacology
9.
Nat Commun ; 11(1): 4684, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32943635

ABSTRACT

Cancer cells have a characteristic metabolism, mostly caused by alterations in signal transduction networks rather than mutations in metabolic enzymes. For metabolic drugs to be cancer-selective, signaling alterations need to be identified that confer a druggable vulnerability. Here, we demonstrate that many tumor cells with an acquired cancer drug resistance exhibit increased sensitivity to mechanistically distinct inhibitors of cancer metabolism. We demonstrate that this metabolic vulnerability is driven by mTORC1, which promotes resistance to chemotherapy and targeted cancer drugs, but simultaneously suppresses autophagy. We show that autophagy is essential for tumor cells to cope with therapeutic perturbation of metabolism and that mTORC1-mediated suppression of autophagy is required and sufficient for generating a metabolic vulnerability leading to energy crisis and apoptosis. Our study links mTOR-induced cancer drug resistance to autophagy defects as a cause of a metabolic liability and opens a therapeutic window for the treatment of otherwise therapy-refractory tumor patients.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , TOR Serine-Threonine Kinases/metabolism , Animals , Apoptosis/drug effects , Autophagy/genetics , Cell Line, Tumor , Cell Survival/drug effects , Deoxyglucose , Drug Therapy , Female , Humans , Lung Neoplasms , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
10.
EMBO J ; 38(20): e102096, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31483066

ABSTRACT

Engineered p53 mutant mice are valuable tools for delineating p53 functions in tumor suppression and cancer therapy. Here, we have introduced the R178E mutation into the Trp53 gene of mice to specifically ablate the cooperative nature of p53 DNA binding. Trp53R178E mice show no detectable target gene regulation and, at first sight, are largely indistinguishable from Trp53-/- mice. Surprisingly, stabilization of p53R178E in Mdm2-/- mice nevertheless triggers extensive apoptosis, indicative of residual wild-type activities. Although this apoptotic activity suffices to trigger lethality of Trp53R178E ;Mdm2-/- embryos, it proves insufficient for suppression of spontaneous and oncogene-driven tumorigenesis. Trp53R178E mice develop tumors indistinguishably from Trp53-/- mice and tumors retain and even stabilize the p53R178E protein, further attesting to the lack of significant tumor suppressor activity. However, Trp53R178E tumors exhibit remarkably better chemotherapy responses than Trp53-/- ones, resulting in enhanced eradication of p53-mutated tumor cells. Together, this provides genetic proof-of-principle evidence that a p53 mutant can be highly tumorigenic and yet retain apoptotic activity which provides a survival benefit in the context of cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Leukemia, Myeloid, Acute/prevention & control , Lymphoma/prevention & control , Mutation , Proto-Oncogene Proteins c-mdm2/physiology , Tumor Suppressor Protein p53/physiology , Animals , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Cycle , Disease Models, Animal , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Lymphoma/genetics , Lymphoma/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Cells, Cultured
11.
Proc Natl Acad Sci U S A ; 113(52): E8433-E8442, 2016 12 27.
Article in English | MEDLINE | ID: mdl-27956623

ABSTRACT

Mutations in the p53 tumor suppressor gene are the most frequent genetic alteration in cancer and are often associated with progression from benign to invasive stages with metastatic potential. Mutations inactivate tumor suppression by p53, and some endow the protein with novel gain of function (GOF) properties that actively promote tumor progression and metastasis. By comparative gene expression profiling of p53-mutated and p53-depleted cancer cells, we identified ectonucleoside triphosphate diphosphohydrolase 5 (ENTPD5) as a mutant p53 target gene, which functions as a uridine 5'-diphosphatase (UDPase) in the endoplasmic reticulum (ER) to promote the folding of N-glycosylated membrane proteins. A comprehensive pan-cancer analysis revealed a highly significant correlation between p53 GOF mutations and ENTPD5 expression. Mechanistically, mutp53 is recruited by Sp1 to the ENTPD5 core promoter to induce its expression. We show ENTPD5 to be a mediator of mutant p53 GOF activity in clonogenic growth, architectural tissue remodeling, migration, invasion, and lung colonization in an experimental metastasis mouse model. Our study reveals folding of N-glycosylated membrane proteins in the ER as a mechanism underlying the metastatic progression of tumors with mutp53 that could provide new possibilities for cancer treatment.


Subject(s)
Endoplasmic Reticulum/metabolism , Neoplasm Metastasis , Oncogene Proteins/metabolism , Pyrophosphatases/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology , Animals , Apoptosis , Calnexin/metabolism , Calreticulin/metabolism , Carcinogenesis/metabolism , Cell Line, Tumor , Disease Progression , Female , Glycoproteins/metabolism , Glycosylation , Humans , Male , Mice , Mutant Proteins/genetics , Mutant Proteins/physiology , Mutation , Neoplasm Invasiveness , Prognosis , RNA Interference , RNA, Small Interfering/metabolism , Sp1 Transcription Factor/metabolism
12.
Nucleic Acids Res ; 44(7): 3204-18, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-26819410

ABSTRACT

TP63, a member of the p53 gene family gene, encodes the ΔNp63 protein and is one of the most frequently amplified genes in squamous cell carcinomas (SCC) of the head and neck (HNSCC) and lungs (LUSC). Using an epiallelic series of siRNAs with intrinsically different knockdown abilities, we show that the complete loss of ΔNp63 strongly impaired cell proliferation, whereas partial ΔNp63 depletion rendered cells hypersensitive to cisplatin accompanied by an accumulation of DNA damage. Expression profiling revealed wide-spread transcriptional regulation of DNA repair genes and in particular Fanconi anemia (FA) pathway components such as FANCD2 and RAD18 - known to be crucial for the repair of cisplatin-induced interstrand crosslinks. In SCC patients ΔNp63 levels significantly correlate with FANCD2 and RAD18 expression confirming ΔNp63 as a key activator of the FA pathway in vivo Mechanistically, ΔNp63 bound an upstream enhancer of FANCD2 inactive in primary keratinocytes but aberrantly activated by ΔNp63 in SCC. Consistently, depletion of FANCD2 sensitized to cisplatin similar to depletion of ΔNp63. Together, our results demonstrate that ΔNp63 directly activates the FA pathway in SCC and limits the efficacy of cisplatin treatment. Targeting ΔNp63 therefore would not only inhibit SCC proliferation but also sensitize tumors to chemotherapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Squamous Cell/genetics , Cisplatin/therapeutic use , DNA Repair , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cells, Cultured , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm , Enhancer Elements, Genetic , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Humans , Transcription Factors/physiology , Transcriptional Activation , Tumor Suppressor Proteins/physiology , Ubiquitin-Protein Ligases/metabolism
13.
Nat Chem Biol ; 12(1): 22-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26595461

ABSTRACT

Inactivation of the p53 tumor suppressor by Mdm2 is one of the most frequent events in cancer, so compounds targeting the p53-Mdm2 interaction are promising for cancer therapy. Mechanisms conferring resistance to p53-reactivating compounds are largely unknown. Here we show using CRISPR-Cas9-based target validation in lung and colorectal cancer that the activity of nutlin, which blocks the p53-binding pocket of Mdm2, strictly depends on functional p53. In contrast, sensitivity to the drug RITA, which binds the Mdm2-interacting N terminus of p53, correlates with induction of DNA damage. Cells with primary or acquired RITA resistance display cross-resistance to DNA crosslinking compounds such as cisplatin and show increased DNA cross-link repair. Inhibition of FancD2 by RNA interference or pharmacological mTOR inhibitors restores RITA sensitivity. The therapeutic response to p53-reactivating compounds is therefore limited by compound-specific resistance mechanisms that can be resolved by CRISPR-Cas9-based target validation and should be considered when allocating patients to p53-reactivating treatments.


Subject(s)
CRISPR-Cas Systems , Drug Resistance, Neoplasm/drug effects , Furans/pharmacology , Genes, p53 , Molecular Targeted Therapy/methods , Cisplatin/pharmacology , DNA Damage/drug effects , DNA Damage/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm/genetics , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Gene Expression Regulation , Genes, p53/physiology , HCT116 Cells/drug effects , Humans , Morpholines/pharmacology , Proto-Oncogene Proteins c-mdm2/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases
14.
Nat Commun ; 5: 3981, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24889111

ABSTRACT

Tumours are heterogeneous cell populations that undergo clonal evolution during tumour progression, metastasis and response to therapy. Short hairpin RNAs (shRNAs) generate stable loss-of-function phenotypes and are versatile experimental tools to explore the contribution of individual genetic alterations to clonal evolution. In these experiments tumour cells carrying shRNAs are commonly tracked with fluorescent reporters. While this works well for cell culture studies and leukaemia mouse models, fluorescent reporters are poorly suited for animals with solid tumours--the most common tumour types in cancer patients. Here we develop a toolkit that uses secreted luciferases to track the fate of two different shRNA-expressing tumour cell clones competitively, both in vitro and in vivo. We demonstrate that secreted luciferase activities can be measured robustly in the blood stream of tumour-bearing mice to accurately quantify, in a minimally invasive manner, the dynamic evolution of two genetically distinct tumour subclones in preclinical mouse models of tumour development, metastasis and therapy.


Subject(s)
Clonal Evolution/genetics , Genetic Vectors , Luciferases , Neoplasms/genetics , RNA, Small Interfering/genetics , Animals , Cell Line, Tumor , Cell Proliferation , HCT116 Cells , Humans , In Vitro Techniques , Mice , Microscopy, Fluorescence , Neoplasms/metabolism
15.
Cell Rep ; 3(5): 1512-25, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23665223

ABSTRACT

Four molecules of the tumor suppressor p53 assemble to cooperatively bind proapoptotic target genes. The structural basis for cooperativity consists of interactions between adjacent DNA binding domains. Mutations at the interaction interface that compromise cooperativity were identified in cancer patients, suggesting a requirement of cooperativity for tumor suppression. We report on an analysis of cooperativity mutant p53E177R mice. Apoptotic functions of p53 triggered by DNA damage and oncogenes were abolished in these mice, whereas functions in cell-cycle control, senescence, metabolism, and antioxidant defense were retained and were sufficient to suppress development of spontaneous T cell lymphoma. Cooperativity mutant mice are nevertheless highly cancer prone and susceptible to different oncogene-induced tumors. Our data underscore the relevance of DNA binding cooperativity for p53-dependent apoptosis and tumor suppression and highlight cooperativity mutations as a class of p53 mutations that result in a selective loss of apoptotic functions due to an altered quaternary structure of the p53 tetramer.


Subject(s)
Apoptosis , DNA/metabolism , Tumor Suppressor Protein p53/metabolism , Aging , Alleles , Animals , Cell Cycle Checkpoints , Cell Transformation, Neoplastic , Cells, Cultured , DNA Damage , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Knock-In Techniques , Germ-Line Mutation , Heterozygote , Lymphoma/etiology , Mice , Mice, Knockout , Protein Binding , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
16.
J Cell Biol ; 188(6): 905-18, 2010 Mar 22.
Article in English | MEDLINE | ID: mdl-20308430

ABSTRACT

Oncogenic stress induces expression of the alternate reading frame (Arf) tumor suppressor protein. Arf then stabilizes p53, which leads to cell cycle arrest or apoptosis. The mechanisms that distinguish both outcomes are incompletely understood. In this study, we show that Arf interacts with the Myc-associated zinc finger protein Miz1. Binding of Arf disrupts the interaction of Miz1 with its coactivator, nucleophosmin, induces the sumoylation of Miz1, and facilitates the assembly of a heterochromatic complex that contains Myc and trimethylated H3K9 in addition to Miz1. Arf-dependent assembly of this complex leads to the repression of multiple genes involved in cell adhesion and signal transduction and induces apoptosis. Our data point to a tumor-suppressive pathway that weakens cell-cell and cell-matrix interactions in response to expression of Arf and that may thereby facilitate the elimination of cells harboring an oncogenic mutation.


Subject(s)
Apoptosis , Kruppel-Like Transcription Factors/metabolism , Tumor Suppressor Protein p14ARF/metabolism , Cell Adhesion , Cells, Cultured , Humans
17.
Blood ; 113(26): 6567-71, 2009 Jun 25.
Article in English | MEDLINE | ID: mdl-19389879

ABSTRACT

Acute myeloid leukemia (AML) patients with internal tandem duplication (ITD) mutations in the Fms-like tyrosine-3 (FLT3) gene have a dismal prognosis. Here we report compassionate-use results with the multikinase and FLT3-ITD inhibitor sorafenib for the treatment of relapsed or refractory FLT3-ITD-positive AML. Sorafenib induced clinically meaningful and very rapid responses in all 6 patients treated either before (n = 2), after (n = 3), or both before and after (n = 1) allogeneic stem cell transplantation (allo-SCT). Sorafenib-induced remissions facilitated allo-SCT in 2 of the 3 refractory patients. Two of the 4 patients who were treated after allo-SCT survived 216 and 221 days, respectively, whereas the other 2 remain in ongoing complete molecular remission. Sorafenib response was associated with an inhibition of the antiapoptotic FLT3-ITD target Stat-5 in vivo. Together, sorafenib monotherapy before or after allo-SCT has remarkable clinical activity in poor risk FLT3-ITD-positive AML and deserves further evaluation in prospective clinical trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzenesulfonates/therapeutic use , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Acute Disease , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Combined Modality Therapy , Drug Evaluation , Drug Resistance, Neoplasm , Female , Humans , Leukemia, Myeloid/surgery , Male , Middle Aged , Neoplasm Proteins/genetics , Niacinamide/analogs & derivatives , Phenylurea Compounds , Remission Induction , Sorafenib , Tandem Repeat Sequences , Transplantation, Homologous , Treatment Outcome , fms-Like Tyrosine Kinase 3/genetics
18.
Nat Cell Biol ; 10(9): 1051-61, 2008 Sep.
Article in English | MEDLINE | ID: mdl-19160485

ABSTRACT

The Myc-associated zinc-finger protein, Miz1, is a negative regulator of cell proliferation and induces expression of the cell-cycle inhibitors p15(Ink4b) and p21(Cip1). Here we identify the ribosomal protein L23 as a negative regulator of Miz1-dependent transactivation. L23 exerts this function by retaining nucleophosmin, an essential co-activator of Miz1 required for Miz1-induced cell-cycle arrest, in the nucleolus. Mutant forms of nucleophosmin found in acute myeloid leukaemia fail to co-activate Miz1 and re-localize it to the cytosol. As L23 is encoded by a direct target gene of Myc, this regulatory circuit may provide a feedback mechanism that links translation of Myc target genes and cell growth to Miz1-dependent cell-cycle arrest.


Subject(s)
Nuclear Proteins/metabolism , Protein Inhibitors of Activated STAT/metabolism , Ribosomal Proteins/metabolism , Alleles , Animals , Cell Proliferation , Feedback, Physiological , HeLa Cells , Humans , Models, Biological , Mutant Proteins/metabolism , Nucleophosmin , Protein Inhibitors of Activated STAT/antagonists & inhibitors , Protein Inhibitors of Activated STAT/chemistry , Protein Structure, Tertiary , Proto-Oncogene Proteins c-myc/metabolism , Rats
19.
Nat Cell Biol ; 9(7): 765-74, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17558397

ABSTRACT

The MYC proto-oncogene encodes a transcription factor that has been implicated in the genesis of many human tumours. Here, we used a bar-code short hairpin RNA (shRNA) screen to identify multiple genes that are required for MYC function. One of these genes encodes USP28, an ubiquitin-specific protease. USP28 is required for MYC stability in human tumour cells. USP28 binds to MYC through an interaction with FBW7alpha, an F-box protein that is part of an SCF-type ubiquitin ligase. Therefore, it stabilizes MYC in the nucleus, but not in the nucleolus, where MYC is degraded by FBW7gamma. High expression levels of USP28 are found in colon and breast carcinomas, and stabilization of MYC by USP28 is essential for tumour-cell proliferation.


Subject(s)
Cell Nucleus/metabolism , F-Box Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitin Thiolesterase/physiology , Adenocarcinoma/metabolism , Adenocarcinoma/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , Cell Nucleolus/metabolism , Cell Proliferation , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Proto-Oncogene Mas
20.
Nat Cell Biol ; 7(1): 30-41, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15580267

ABSTRACT

The transcription factor Miz1 is required for DNA-damage-induced cell-cycle arrest. We have now identified 14-3-3eta as a gene that inhibits Miz1 function through interaction with its DNA binding domain. Binding of 14-3-3eta to Miz1 depends on phosphorylation by Akt and regulates the recovery of cells from arrest after DNA damage. Miz1 has two functions in response to DNA damage: first, it is required for upregulation of a large group of genes, a function that is regulated by c-Myc, but not by 14-3-3eta; second, Miz1 represses the expression of many genes in response to DNA damage in an Akt- and 14-3-3eta-regulated manner.


Subject(s)
14-3-3 Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle/physiology , DNA Damage/genetics , DNA-Binding Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Transcription Factors/metabolism , 14-3-3 Proteins/genetics , Animals , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Gene Expression Regulation/physiology , Gene Library , HeLa Cells , Humans , Kruppel-Like Transcription Factors , Mice , Molecular Sequence Data , NIH 3T3 Cells , Phosphorylation , Protein Binding/physiology , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Rats , Transcription Factors/genetics , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...