Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Cells ; 12(9)2023 04 25.
Article in English | MEDLINE | ID: mdl-37174648

ABSTRACT

Peripheral neuropathy is a common side effect of cancer treatment with paclitaxel. The mechanisms by which paclitaxel is transported into neurons, which are essential for preventing neuropathy, are not well understood. We studied the uptake mechanisms of paclitaxel into neurons using inhibitors for endocytosis, autophagy, organic anion-transporting polypeptide (OATP) drug transporters, and derivatives of paclitaxel. RT-qPCR was used to investigate the expression levels of OATPs in different neuronal tissues and cell lines. OATP transporters were pharmacologically inhibited or modulated by overexpression and CRISPR/Cas9-knock-out to investigate paclitaxel transport in neurons. Through these experiments, we identified OATP1A1 and OATP1B2 as the primary neuronal transporters for paclitaxel. In vitro inhibition of OATP1A1 and OAT1B2 by glycyrrhizic acid attenuated neurotoxicity, while paclitaxel's antineoplastic effects were sustained in cancer cell lines. In vivo, glycyrrhizic acid prevented paclitaxel-induced toxicity and improved behavioral and electrophysiological measures. This study indicates that a set of OATPs are involved in paclitaxel transport into neurons. The inhibition of OATP1A1 and OATP1B2 holds a promising strategy to prevent paclitaxel-induced peripheral neuropathy.


Subject(s)
Organic Anion Transporters , Peripheral Nervous System Diseases , Humans , Paclitaxel/adverse effects , Glycyrrhizic Acid/pharmacology , Organic Anion Transporters/metabolism , Neurons/metabolism , Membrane Transport Proteins , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/prevention & control
2.
Pain ; 164(8): 1718-1733, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-36727909

ABSTRACT

ABSTRACT: Induced pluripotent stem cells (iPSCs) have enabled the generation of various difficult-to-access cell types such as human nociceptors. A key challenge associated with human iPSC-derived nociceptors (hiPSCdNs) is their prolonged functional maturation. While numerous studies have addressed the expression of classic neuronal markers and ion channels in hiPSCdNs, the temporal development of key signaling cascades regulating nociceptor activity has remained largely unexplored. In this study, we used an immunocytochemical high-content imaging approach alongside electrophysiological staging to assess metabotropic and ionotropic signaling of large scale-generated hiPSCdNs across 70 days of in vitro differentiation. During this period, the resting membrane potential became more hyperpolarized, while rheobase, action potential peak amplitude, and membrane capacitance increased. After 70 days, hiPSCdNs exhibited robust physiological responses induced by GABA, pH shift, ATP, and capsaicin. Direct activation of protein kinase A type II (PKA-II) through adenylyl cyclase stimulation with forskolin resulted in PKA-II activation at all time points. Depolarization-induced activation of PKA-II emerged after 35 days of differentiation. However, effective inhibition of forskolin-induced PKA-II activation by opioid receptor agonists required 70 days of in vitro differentiation. Our results identify a pronounced time difference between early expression of functionally important ion channels and emergence of regulatory metabotropic sensitizing and desensitizing signaling only at advanced stages of in vitro cultivation, suggesting an independent regulation of ionotropic and metabotropic signaling. These data are relevant for devising future studies into the development and regulation of human nociceptor function and for defining time windows suitable for hiPSCdN-based drug discovery.


Subject(s)
Induced Pluripotent Stem Cells , Humans , Analgesics, Opioid , Colforsin/pharmacology , Nociception , Sensory Receptor Cells , Ion Channels
3.
Breast Cancer Res ; 24(1): 41, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35715861

ABSTRACT

BACKGROUND: The majority of BRCA1-mutant breast cancers are characterized by a triple-negative phenotype and a basal-like molecular subtype, associated with aggressive clinical behavior. Current treatment options are limited, highlighting the need for the development of novel targeted therapies for this tumor subtype. METHODS: Our group previously showed that EZH2 is functionally relevant in BRCA1-deficient breast tumors and blocking EZH2 enzymatic activity could be a potent treatment strategy. To validate the role of EZH2 as a therapeutic target and to identify new synergistic drug combinations, we performed a high-throughput drug combination screen in various cell lines derived from BRCA1-deficient and -proficient mouse mammary tumors. RESULTS: We identified the combined inhibition of EZH2 and the proximal DNA damage response kinase ATM as a novel synthetic lethality-based therapy for the treatment of BRCA1-deficient breast tumors. We show that the combined treatment with the EZH2 inhibitor GSK126 and the ATM inhibitor AZD1390 led to reduced colony formation, increased genotoxic stress, and apoptosis-mediated cell death in BRCA1-deficient mammary tumor cells in vitro. These findings were corroborated by in vivo experiments showing that simultaneous inhibition of EZH2 and ATM significantly increased anti-tumor activity in mice bearing BRCA1-deficient mammary tumors. CONCLUSION: Taken together, we identified a synthetic lethal interaction between EZH2 and ATM and propose this synergistic interaction as a novel molecular combination for the treatment of BRCA1-mutant breast cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Ataxia Telangiectasia Mutated Proteins , BRCA1 Protein , Breast Neoplasms , Enhancer of Zeste Homolog 2 Protein , Indoles , Protein Kinase Inhibitors , Pyridones , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/deficiency , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Humans , Indoles/pharmacology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Protein Kinase Inhibitors/pharmacology , Pyridones/pharmacology , Synthetic Lethal Mutations
4.
Nat Neurosci ; 25(2): 168-179, 2022 02.
Article in English | MEDLINE | ID: mdl-34931070

ABSTRACT

Bacterial products can act on neurons to alter signaling and function. In the present study, we found that dorsal root ganglion (DRG) sensory neurons are enriched for ANTXR2, the high-affinity receptor for anthrax toxins. Anthrax toxins are composed of protective antigen (PA), which binds to ANTXR2, and the protein cargoes edema factor (EF) and lethal factor (LF). Intrathecal administration of edema toxin (ET (PA + EF)) targeted DRG neurons and induced analgesia in mice. ET inhibited mechanical and thermal sensation, and pain caused by formalin, carrageenan or nerve injury. Analgesia depended on ANTXR2 expressed by Nav1.8+ or Advillin+ neurons. ET modulated protein kinase A signaling in mouse sensory and human induced pluripotent stem cell-derived sensory neurons, and attenuated spinal cord neurotransmission. We further engineered anthrax toxins to introduce exogenous protein cargoes, including botulinum toxin, into DRG neurons to silence pain. Our study highlights interactions between a bacterial toxin and nociceptors, which may lead to the development of new pain therapeutics.


Subject(s)
Anthrax , Bacillus anthracis , Bacterial Toxins , Induced Pluripotent Stem Cells , Animals , Anthrax/microbiology , Anthrax/therapy , Bacillus anthracis/metabolism , Bacterial Toxins/metabolism , Ganglia, Spinal/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Nociceptors/metabolism , Pain , Receptors, Peptide/metabolism
5.
Sci Transl Med ; 13(619): eabh1314, 2021 11 10.
Article in English | MEDLINE | ID: mdl-34757807

ABSTRACT

The voltage-gated sodium NaV1.7 channel, critical for sensing pain, has been actively targeted by drug developers; however, there are currently no effective and safe therapies targeting NaV1.7. Here, we tested whether a different approach, indirect NaV1.7 regulation, could have antinociceptive effects in preclinical models. We found that preventing addition of small ubiquitin-like modifier (SUMO) on the NaV1.7-interacting cytosolic collapsin response mediator protein 2 (CRMP2) blocked NaV1.7 functions and had antinociceptive effects in rodents. In silico targeting of the SUMOylation site in CRMP2 (Lys374) identified >200 hits, of which compound 194 exhibited selective in vitro and ex vivo NaV1.7 engagement. Orally administered 194 was not only antinociceptive in preclinical models of acute and chronic pain but also demonstrated synergy alongside other analgesics­without eliciting addiction, rewarding properties, or neurotoxicity. Analgesia conferred by 194 was opioid receptor dependent. Our results demonstrate that 194 is a first-in-class protein-protein inhibitor that capitalizes on CRMP2-NaV1.7 regulation to deliver safe analgesia in rodents.


Subject(s)
Chronic Pain , NAV1.7 Voltage-Gated Sodium Channel , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Rodentia/metabolism , Sumoylation
6.
Hum Genet ; 140(12): 1679-1693, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34545459

ABSTRACT

The highly conserved YrdC domain-containing protein (YRDC) interacts with the well-described KEOPS complex, regulating specific tRNA modifications to ensure accurate protein synthesis. Previous studies have linked the KEOPS complex to a role in promoting telomere maintenance and controlling genome integrity. Here, we report on a newborn with a severe neonatal progeroid phenotype including generalized loss of subcutaneous fat, microcephaly, growth retardation, wrinkled skin, renal failure, and premature death at the age of 12 days. By trio whole-exome sequencing, we identified a novel homozygous missense mutation, c.662T > C, in YRDC affecting an evolutionary highly conserved amino acid (p.Ile221Thr). Functional characterization of patient-derived dermal fibroblasts revealed that this mutation impairs YRDC function and consequently results in reduced t6A modifications of tRNAs. Furthermore, we established and performed a novel and highly sensitive 3-D Q-FISH analysis based on single-telomere detection to investigate the impact of YRDC on telomere maintenance. This analysis revealed significant telomere shortening in YRDC-mutant cells. Moreover, single-cell RNA sequencing analysis of YRDC-mutant fibroblasts revealed significant transcriptome-wide changes in gene expression, specifically enriched for genes associated with processes involved in DNA repair. We next examined the DNA damage response of patient's dermal fibroblasts and detected an increased susceptibility to genotoxic agents and a global DNA double-strand break repair defect. Thus, our data suggest that YRDC may affect the maintenance of genomic stability. Together, our findings indicate that biallelic variants in YRDC result in a developmental disorder with progeroid features and might be linked to increased genomic instability and telomere shortening.


Subject(s)
Developmental Disabilities/genetics , GTP-Binding Proteins/genetics , Progeria/genetics , RNA-Binding Proteins/genetics , Alleles , Consanguinity , DNA Damage , Developmental Disabilities/pathology , Genome, Human , Genomic Instability , Homozygote , Humans , Infant, Newborn , Male , Mutation , Pedigree , Progeria/pathology , RNA, Transfer/genetics , Sequence Analysis, RNA , Telomere Shortening
7.
J Cell Biol ; 220(10)2021 10 04.
Article in English | MEDLINE | ID: mdl-34431981

ABSTRACT

Depolarization drives neuronal plasticity. However, whether depolarization drives sensitization of peripheral nociceptive neurons remains elusive. By high-content screening (HCS) microscopy, we revealed that depolarization of cultured sensory neurons rapidly activates protein kinase A type II (PKA-II) in nociceptors by calcium influx through CaV1.2 channels. This effect was modulated by calpains but insensitive to inhibitors of cAMP formation, including opioids. In turn, PKA-II phosphorylated Ser1928 in the distal C terminus of CaV1.2, thereby increasing channel gating, whereas dephosphorylation of Ser1928 involved the phosphatase calcineurin. Patch-clamp and behavioral experiments confirmed that depolarization leads to calcium- and PKA-dependent sensitization of calcium currents ex vivo and local peripheral hyperalgesia in the skin in vivo. Our data suggest a local activity-driven feed-forward mechanism that selectively translates strong depolarization into further activity and thereby facilitates hypersensitivity of nociceptor terminals by a mechanism inaccessible to opioids.


Subject(s)
Calcium Channels, L-Type/metabolism , Cyclic AMP-Dependent Protein Kinase Type II/metabolism , Nociceptors/metabolism , Animals , Male , Rats , Rats, Sprague-Dawley
8.
Cell Death Dis ; 12(6): 530, 2021 05 24.
Article in English | MEDLINE | ID: mdl-34031359

ABSTRACT

Non-coding RNA from pericentromeric satellite repeats are involved in stress-dependent splicing processes, maintenance of heterochromatin, and are required to protect genome stability. Here we show that the long non-coding satellite III RNA (SatIII) generates resistance against the topoisomerase IIa (TOP2A) inhibitor etoposide in lung cancer. Because heat shock conditions (HS) protect cells against the toxicity of etoposide, and SatIII is significantly induced under HS, we hypothesized that the protective effect could be traced back to SatIII. Using genome methylation profiles of patient-derived xenograft mouse models we show that the epigenetic modification of the SatIII DNA locus and the resulting SatIII expression predict chemotherapy resistance. In response to stress, SatIII recruits TOP2A to nuclear stress bodies, which protects TOP2A from a complex formation with etoposide and results in decreased DNA damage after treatment. We show that BRD4 inhibitors reduce the expression of SatIII, restoring etoposide sensitivity.


Subject(s)
Drug Resistance, Neoplasm/genetics , Etoposide/therapeutic use , RNA, Long Noncoding/physiology , Animals , Antineoplastic Agents/pharmacology , Cell Cycle Proteins/antagonists & inhibitors , Centromere/genetics , Centromere/metabolism , DNA Methylation/physiology , DNA Topoisomerases, Type II/drug effects , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HEK293 Cells , HeLa Cells , Humans , Male , Mice, Inbred NOD , Mice, SCID , Poly-ADP-Ribose Binding Proteins/drug effects , Poly-ADP-Ribose Binding Proteins/genetics , Poly-ADP-Ribose Binding Proteins/metabolism , RNA, Long Noncoding/genetics , Transcription Factors/antagonists & inhibitors , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
FEBS J ; 288(7): 2418-2435, 2021 04.
Article in English | MEDLINE | ID: mdl-33051988

ABSTRACT

Venoms are a rich source of highly specific toxins, which allow the identification of novel therapeutic targets. We have now applied high content screening (HCS) microscopy to identify toxins that modulate pain sensitization signaling in primary sensory neurons of rat and elucidated the underlying mechanism. A set of venoms and fractions thereof were analyzed for their ability to activate type II protein kinase A (PKA-II) and extracellular signal-regulated kinases (ERK1/2). We identified MeuNaTxα-1, a sodium channel-selective scorpion α-toxin from Mesobuthus eupeus, which affected both PKA-II and ERK1/2. Recombinant MeuNaTxα-1 showed identical activity to the native toxin on mammalian voltage-gated sodium channels expressed in Xenopus laevis oocytes and induced thermal hyperalgesia in adult mice. The effect of MeuNaTxα-1 on sensory neurons was dose-dependent and tetrodotoxin-sensitive. Application of inhibitors and toxin mutants with altered sodium channel selectivity demonstrated that signaling activation in sensory neurons depends on NaV 1.2 isoform. Accordingly, the toxin was more potent in neurons from newborn rats, where NaV 1.2 is expressed at a higher level. Our results demonstrate that HCS microscopy-based monitoring of intracellular signaling is a novel and powerful tool to identify and characterize venoms and their toxins affecting sensory neurons.


Subject(s)
Cyclic AMP-Dependent Protein Kinase Type II/genetics , NAV1.2 Voltage-Gated Sodium Channel/genetics , Pain/genetics , Voltage-Gated Sodium Channels/genetics , Animals , Animals, Newborn , Humans , Hyperalgesia/genetics , Hyperalgesia/pathology , MAP Kinase Signaling System/drug effects , Mice , Oocytes/drug effects , Oocytes/growth & development , Rats , Scorpion Venoms/chemistry , Scorpion Venoms/pharmacology , Scorpions/chemistry , Sensory Receptor Cells , Xenopus laevis/growth & development
10.
Sci Rep ; 9(1): 16272, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31700036

ABSTRACT

The α2δ-1 subunit of voltage-gated calcium channels binds to gabapentin and pregabalin, mediating the analgesic action of these drugs against neuropathic pain. Extracellular matrix proteins from the thrombospondin (TSP) family have been identified as ligands of α2δ-1 in the CNS. This interaction was found to be crucial for excitatory synaptogenesis and neuronal sensitisation which in turn can be inhibited by gabapentin, suggesting a potential role in the pathogenesis of neuropathic pain. Here, we provide information on the biochemical properties of the direct TSP/α2δ-1 interaction using an ELISA-style ligand binding assay. Our data reveal that full-length pentameric TSP-4, but neither TSP-5/COMP of the pentamer-forming subgroup B nor TSP-2 of the trimer-forming subgroup A directly interact with a soluble variant of α2δ-1 (α2δ-1S). Interestingly, this interaction is not inhibited by gabapentin on a molecular level and is not detectable on the surface of HEK293-EBNA cells over-expressing α2δ-1 protein. These results provide biochemical evidence that supports a specific role of TSP-4 among the TSPs in mediating the binding to neuronal α2δ-1 and suggest that gabapentin does not directly target TSP/α2δ-1 interaction to alleviate neuropathic pain.


Subject(s)
Calcium Channels, L-Type/metabolism , Gabapentin/metabolism , Thrombospondins/metabolism , Enzyme-Linked Immunosorbent Assay , HEK293 Cells , Humans , Ligands , Mass Spectrometry , Membrane Proteins/chemistry , Membrane Proteins/metabolism
11.
Cancer Res ; 79(19): 4855-4868, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31405847

ABSTRACT

The advent of molecularly targeted therapeutic agents has opened a new era in cancer therapy. However, many tumors rely on nondruggable cancer-driving lesions. In addition, long-lasting clinical benefits from single-agent therapies rarely occur, as most of the tumors acquire resistance over time. The identification of targeted combination regimens interfering with signaling through oncogenically rewired pathways provides a promising approach to enhance efficacy of single-agent-targeted treatments. Moreover, combination drug therapies might overcome the emergence of drug resistance. Here, we performed a focused flow cytometry-based drug synergy screen and identified a novel synergistic interaction between GLUT1-mediated glucose transport and the cell-cycle checkpoint kinases ATR and CHK1. Combined inhibition of CHK1/GLUT1 or ATR/GLUT1 robustly induced apoptosis, particularly in RAS-mutant cancer cells. Mechanistically, combined inhibition of ATR/CHK1 and GLUT1 arrested sensitive cells in S-phase and led to the accumulation of genotoxic damage, particularly in S-phase. In vivo, simultaneous inhibition of ATR and GLUT1 significantly reduced tumor volume gain in an autochthonous mouse model of KrasG12D -driven soft tissue sarcoma. Taken together, these findings pave the way for combined inhibition of GLUT1 and ATR/CHK1 as a therapeutic approach for KRAS-driven cancers. SIGNIFICANCE: Dual targeting of the DNA damage response and glucose transport synergistically induces apoptosis in KRAS-mutant cancer, suggesting this combination treatment for clinical validation in KRAS-stratified tumor patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Checkpoint Kinase 1/antagonists & inhibitors , Glucose Transporter Type 1/antagonists & inhibitors , Neoplasms, Experimental/pathology , Animals , Apoptosis/drug effects , Benzodiazepinones/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Drug Synergism , Humans , Hydroxybenzoates/pharmacology , Mice , Mice, Nude , Molecular Targeted Therapy/methods , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Pyrazoles/pharmacology , Quinazolines/pharmacology , Xenograft Model Antitumor Assays
12.
Methods Mol Biol ; 1987: 111-124, 2019.
Article in English | MEDLINE | ID: mdl-31028677

ABSTRACT

Studying TRP channel expressing nociceptors requires the identification of the respective subpopulations as well as the quantification of dynamic cellular events. However, the heterogeneity of sensory neurons and associated nonneuronal cells demands the analysis of large numbers of cells to reflect the distribution of entire populations. Here we report a detailed workflow how to apply high-content screening (HCS) microscopy to signaling events in TRPV1-positive neurons as well as an approach to use the selective elimination of TRPV1 positive cells from dissociated rat sensory ganglia as base for transcriptomic analysis of TRPV1-positive cells and/or as control for TRPV1 antibody specificity.


Subject(s)
Sensory Receptor Cells/metabolism , TRPV Cation Channels/metabolism , Animals , Cells, Cultured , Fluorescent Antibody Technique/methods , Male , Mice, Inbred C57BL , Microscopy/methods , Microscopy, Fluorescence , Nociceptors/metabolism , Rats , Rats, Sprague-Dawley , TRPV Cation Channels/immunology
13.
Cell ; 176(3): 505-519.e22, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30612738

ABSTRACT

Genomic instability can be a hallmark of both human genetic disease and cancer. We identify a deleterious UBQLN4 mutation in families with an autosomal recessive syndrome reminiscent of genome instability disorders. UBQLN4 deficiency leads to increased sensitivity to genotoxic stress and delayed DNA double-strand break (DSB) repair. The proteasomal shuttle factor UBQLN4 is phosphorylated by ATM and interacts with ubiquitylated MRE11 to mediate early steps of homologous recombination-mediated DSB repair (HRR). Loss of UBQLN4 leads to chromatin retention of MRE11, promoting non-physiological HRR activity in vitro and in vivo. Conversely, UBQLN4 overexpression represses HRR and favors non-homologous end joining. Moreover, we find UBQLN4 overexpressed in aggressive tumors. In line with an HRR defect in these tumors, UBQLN4 overexpression is associated with PARP1 inhibitor sensitivity. UBQLN4 therefore curtails HRR activity through removal of MRE11 from damaged chromatin and thus offers a therapeutic window for PARP1 inhibitor treatment in UBQLN4-overexpressing tumors.


Subject(s)
Carrier Proteins/genetics , Nuclear Proteins/genetics , Carrier Proteins/metabolism , Chromatin/metabolism , DNA , DNA Breaks, Double-Stranded , DNA Damage/genetics , DNA End-Joining Repair , DNA-Binding Proteins/metabolism , Female , Genomic Instability , Germ-Line Mutation , Homologous Recombination , Humans , MRE11 Homologue Protein/genetics , MRE11 Homologue Protein/metabolism , Male , Neoplasms/genetics , Neoplasms/metabolism , Nuclear Proteins/metabolism , Primary Cell Culture , Recombinational DNA Repair
14.
Cell Rep ; 25(4): 1027-1039.e6, 2018 10 23.
Article in English | MEDLINE | ID: mdl-30355482

ABSTRACT

Cdkn1a, which encodes p21, functions as a major route for p53-mediated cell-cycle arrest. However, the consequence of Cdkn1a gene dosage on tumor suppression has not been systematically investigated. Here, we employed BAC transgenesis to generate a Cdkn1aSUPER mouse, which harbors an additional Cdkn1a allele within its natural genomic context. We show that these mice display enhanced cell-cycle arrest and reduced apoptosis in response to genotoxic stress. Furthermore, using a chemically induced skin cancer model and an autochthonous Kras-driven lung adenocarcinoma model, we show that Cdkn1aSUPER mice display a cancer protection phenotype that is indistinguishable from that observed in Tp53SUPER animals. Moreover, we demonstrate that Tp53 and Cdkn1a cooperate in mediating cancer resistance, using a chemically induced fibrosarcoma model. Overall, our Cdkn1aSUPER allele enabled us to assess the contribution of Cdkn1a to Tp53-mediated tumor suppression.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Carcinogenesis/pathology , Cell Cycle Checkpoints , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cytoprotection , DNA Damage , Drug Resistance, Neoplasm , Embryo, Mammalian/cytology , Epithelium/metabolism , Fibroblasts/metabolism , Gene Dosage , Mice , Mice, Inbred C57BL , Mice, Transgenic , Regeneration
15.
J Cell Biol ; 217(6): 2167-2184, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29615473

ABSTRACT

Type II isoforms of cyclic adenosine monophosphate (cAMP)-dependent protein kinase A (PKA-II) contain a phosphorylatable epitope within the inhibitory domain of RII subunits (pRII) with still unclear function. In vitro, RII phosphorylation occurs in the absence of cAMP, whereas staining of cells with pRII-specific antibodies revealed a cAMP-dependent pattern. In sensory neurons, we found that increased pRII immunoreactivity reflects increased accessibility of the already phosphorylated RII epitope during cAMP-induced opening of the tetrameric RII2:C2 holoenzyme. Accordingly, induction of pRII by cAMP was sensitive to novel inhibitors of dissociation, whereas blocking catalytic activity was ineffective. Also in vitro, cAMP increased the binding of pRII antibodies to RII2:C2 holoenzymes. Identification of an antibody specific for the glycine-rich loop of catalytic subunits facing the pRII-epitope confirmed activity-dependent binding with similar kinetics, proving that the reassociation is rapid and precisely controlled. Mechanistic modeling further supported that RII phosphorylation precedes cAMP binding and controls the inactivation by modulating the reassociation involving the coordinated action of phosphodiesterases and phosphatases.


Subject(s)
Cyclic AMP-Dependent Protein Kinase RIIalpha Subunit/metabolism , Cyclic AMP/metabolism , Protein Subunits/metabolism , Adenosine Triphosphate/metabolism , Animals , Antibodies/pharmacology , Cell Extracts , Cell Membrane Permeability/drug effects , Cyclic AMP-Dependent Protein Kinase RIIalpha Subunit/chemistry , Enzyme Activation/drug effects , HEK293 Cells , Humans , Isoenzymes/metabolism , Male , Mice , Models, Biological , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Structure, Secondary , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Signal Transduction/drug effects
16.
Oncotarget ; 8(27): 44418-44433, 2017 07 04.
Article in English | MEDLINE | ID: mdl-28574843

ABSTRACT

The composition of tumor-infiltrating lymphocytes (TIL) reflects biology and immunogenicity of cancer. Here, we characterize T-cell subsets and expression of immune checkpoint molecules in head and neck squamous cell carcinoma (HNSCC). We analyzed TIL subsets in primary tumors (n = 34), blood (peripheral blood mononuclear cells (PBMC); n = 34) and non-cancerous mucosa (n = 7) of 34 treatment-naïve HNSCC patients and PBMC of 15 healthy controls. Flow cytometry analyses revealed a highly variable T-cell infiltration mainly of an effector memory phenotype (CD45RA-/CCR7-). Naïve T cells (CD45RA+/CCR7+) were decreased in the microenvironment compared to PBMC of patients, while regulatory T cells (CD4+/CD25+/CD127low and CD4+/CD39+) were elevated. Furthermore, we performed digital image analyses of entire cross sections of HNSCC to define the 'Immunoscore' (CD3+ and CD8+ cell infiltration in tumor core and invasive margin) and quantified MHC class I expression on tumor cells by immunohistochemistry. Immune checkpoint molecules cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed cell death 1 (PD-1) and programmed cell death 1 ligand 1 (PD-L1) were increased in TILs compared to peripheral T cells in flow-cytometric analysis. Human papillomavirus (HPV) positive tumors showed higher numbers of TILs, but a similar composition of T-cell subsets and checkpoint molecule expression compared to HPV negative tumors. Taken together, the tumor microenvironment of HNSCC is characterized by a strong infiltration of regulatory T cells and high checkpoint molecule expression on T-cell subsets. In view of increasingly used immunotherapies, a detailed knowledge of TILs and checkpoint molecule expression on TILs is of high translational relevance.


Subject(s)
Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/metabolism , Immunomodulation , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Biomarkers , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Carcinoma, Squamous Cell/pathology , Female , Head and Neck Neoplasms/pathology , Humans , Immunophenotyping , Lymphocyte Count , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Phenotype , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Squamous Cell Carcinoma of Head and Neck , Tumor Microenvironment/immunology
17.
Sci Signal ; 10(461)2017 01 10.
Article in English | MEDLINE | ID: mdl-28074005

ABSTRACT

Genetic loss of the voltage-gated sodium channel Nav1.7 (Nav1.7-/-) results in lifelong insensitivity to pain in mice and humans. One underlying cause is an increase in the production of endogenous opioids in sensory neurons. We analyzed whether Nav1.7 deficiency altered nociceptive heterotrimeric guanine nucleotide-binding protein-coupled receptor (GPCR) signaling, such as initiated by GPCRs that respond to serotonin (pronociceptive) or opioids (antinociceptive), in sensory neurons. We found that the nociceptive neurons of Nav1.7 knockout (Nav1.7-/-) mice, but not those of Nav1.8 knockout (Nav1.8-/-) mice, exhibited decreased pronociceptive serotonergic signaling through the 5-HT4 receptors, which are Gαs-coupled GPCRs that stimulate the production of cyclic adenosine monophosphate resulting in protein kinase A (PKA) activity, as well as reduced abundance of the RIIß regulatory subunit of PKA. Simultaneously, the efficacy of antinociceptive opioid signaling mediated by the Gαi-coupled mu opioid receptors was increased. Consequently, opioids inhibited more efficiently tetrodotoxin-resistant sodium currents, which are important for pain-initiating neuronal activity in nociceptive neurons. Thus, Nav1.7 controls the efficacy and balance of GPCR-mediated pro- and antinociceptive intracellular signaling, such that without Nav1.7, the balance is shifted toward antinociception, resulting in lifelong endogenous analgesia.


Subject(s)
Analgesics, Opioid/metabolism , NAV1.7 Voltage-Gated Sodium Channel/metabolism , Pain/metabolism , Serotonin/metabolism , Signal Transduction , Action Potentials/drug effects , Analgesics, Opioid/pharmacology , Animals , Cyclic AMP-Dependent Protein Kinase RIIbeta Subunit/metabolism , Ganglia, Spinal/metabolism , Indoles/pharmacology , Male , Mice, Knockout , NAV1.7 Voltage-Gated Sodium Channel/genetics , Nociceptors/drug effects , Nociceptors/metabolism , Pain/genetics , Patch-Clamp Techniques , Rats, Sprague-Dawley , Receptors, Opioid, mu/metabolism , Receptors, Serotonin, 5-HT4/metabolism , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Serotonin/pharmacology , Serotonin Antagonists/pharmacology , Sodium Channel Blockers/pharmacology , Sulfonamides/pharmacology , Tetrodotoxin/pharmacology
18.
Nucleic Acids Res ; 45(1): 382-394, 2017 Jan 09.
Article in English | MEDLINE | ID: mdl-27536004

ABSTRACT

The cellular response to heat stress is an ancient and evolutionarily highly conserved defence mechanism characterised by the transcriptional up-regulation of cyto-protective genes and a partial inhibition of splicing. These features closely resemble the proteotoxic stress response during tumor development. The bromodomain protein BRD4 has been identified as an integral member of the oxidative stress as well as of the inflammatory response, mainly due to its role in the transcriptional regulation process. In addition, there are also several lines of evidence implicating BRD4 in the splicing process. Using RNA-sequencing we found a significant increase in splicing inhibition, in particular intron retentions (IR), following heat treatment in BRD4-depleted cells. This leads to a decrease of mRNA abundancy of the affected transcripts, most likely due to premature termination codons. Subsequent experiments revealed that BRD4 interacts with the heat shock factor 1 (HSF1) such that under heat stress BRD4 is recruited to nuclear stress bodies and non-coding SatIII RNA transcripts are up-regulated. These findings implicate BRD4 as an important regulator of splicing during heat stress. Our data which links BRD4 to the stress induced splicing process may provide novel mechanisms of BRD4 inhibitors in regard to anti-cancer therapies.


Subject(s)
DNA-Binding Proteins/genetics , Heat-Shock Response/genetics , Nuclear Proteins/genetics , RNA Splicing , RNA, Messenger/genetics , RNA, Untranslated/genetics , Transcription Factors/genetics , DNA-Binding Proteins/metabolism , Exons , HeLa Cells , Heat Shock Transcription Factors , Histone Acetyltransferases , Histone Chaperones , Hot Temperature , Humans , Introns , Nuclear Proteins/metabolism , Protein Domains , RNA, Messenger/metabolism , RNA, Untranslated/metabolism , Sequence Analysis, RNA , Transcription Factors/metabolism
19.
Nucleic Acids Res ; 42(10): 6436-47, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24728989

ABSTRACT

The antimetabolite 5-fluorouracil is a widely used chemotherapeutic for the treatment of several solid cancers. However, resistance to 5-fluorouracil remains a major drawback in its clinical use. In this study we report that treatment of HeLa cells with 5-fluorouracil resulted in de novo assembly of stress granules. Moreover, we revealed that stress granule assembly under stress conditions as well as disassembly is altered in cells treated with 5-fluorouracil. Notably, we discovered that RACK1, a protein mediating cell survival and apoptosis, is a component of 5-fluorouracil-induced stress granules. To explore the mode of action of 5-fluorouracil accountable for de novo stress granule assembly, we analyzed 5-fluorouracil metabolites and noticed that stress granule assembly is caused by RNA, not DNA incorporating 5-fluorouracil metabolites. Interestingly, we observed that other RNA incorporating drugs also cause assembly of stress granules. Thus, our results suggest that incorporation of chemotherapeutics into RNA may result in stress granule assembly with potential significance in chemoresistance.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Fluorouracil/pharmacology , RNA/metabolism , Ribonucleoproteins/metabolism , Stress, Physiological , Antimetabolites, Antineoplastic/metabolism , Azacitidine/metabolism , Cell Line , DNA/metabolism , Eukaryotic Initiation Factor-2/metabolism , Fluorouracil/metabolism , GTP-Binding Proteins/analysis , HeLa Cells , Humans , Neoplasm Proteins/analysis , Oxidative Stress , Receptors for Activated C Kinase , Receptors, Cell Surface/analysis , Ribonucleoproteins/analysis , Thioguanine/metabolism
20.
Gene ; 540(2): 210-6, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24582972

ABSTRACT

The G protein-coupled receptor 30 (GPR30) has been claimed as an estrogen receptor. However, the literature reports controversial findings and the physiological function of GPR30 is not fully understood yet. Consistent with studies assigning a role of GPR30 in the cardiovascular and metabolic systems, GPR30 expression has been reported in small arterial vessels, pancreas and chief gastric cells of the stomach. Therefore, we hypothesized a role of GPR30 in the onset and progression of cardiovascular and metabolic diseases. In order to test our hypothesis, we investigated the effects of a high-fat diet on the metabolic and cardiovascular profiles of Gpr30-deficient mice (GPR30-lacZ mice). We found that GPR30-lacZ female, rather than male, mice had significant lower levels of HDL along with an increase in fat liver accumulation as compared to control mice. However, two indicators of cardiac performance assessed by echocardiography, ejection fraction and fractional shortening were both decreased in an age-dependent manner only in Gpr30-lacZ male mice. Collectively our results point to a potential role of Gpr30 in preserving lipid metabolism and cardiac function in a sex- and age-dependent fashion.


Subject(s)
Aorta/physiopathology , Heart/physiopathology , Obesity/metabolism , Receptors, G-Protein-Coupled/genetics , Adiposity , Age Factors , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Blood Flow Velocity , Diet, High-Fat/adverse effects , Female , Gene Deletion , Genetic Association Studies , Lipoproteins, HDL/blood , Liver/metabolism , Liver/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Obesity/genetics , Receptors, Estrogen , Receptors, G-Protein-Coupled/deficiency , Sex Characteristics , Stroke Volume
SELECTION OF CITATIONS
SEARCH DETAIL
...