Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
2.
Nature ; 613(7945): 759-766, 2023 01.
Article in English | MEDLINE | ID: mdl-36631611

ABSTRACT

Protein phosphorylation is one of the most widespread post-translational modifications in biology1,2. With advances in mass-spectrometry-based phosphoproteomics, 90,000 sites of serine and threonine phosphorylation have so far been identified, and several thousand have been associated with human diseases and biological processes3,4. For the vast majority of phosphorylation events, it is not yet known which of the more than 300 protein serine/threonine (Ser/Thr) kinases encoded in the human genome are responsible3. Here we used synthetic peptide libraries to profile the substrate sequence specificity of 303 Ser/Thr kinases, comprising more than 84% of those predicted to be active in humans. Viewed in its entirety, the substrate specificity of the kinome was substantially more diverse than expected and was driven extensively by negative selectivity. We used our kinome-wide dataset to computationally annotate and identify the kinases capable of phosphorylating every reported phosphorylation site in the human Ser/Thr phosphoproteome. For the small minority of phosphosites for which the putative protein kinases involved have been previously reported, our predictions were in excellent agreement. When this approach was applied to examine the signalling response of tissues and cell lines to hormones, growth factors, targeted inhibitors and environmental or genetic perturbations, it revealed unexpected insights into pathway complexity and compensation. Overall, these studies reveal the intrinsic substrate specificity of the human Ser/Thr kinome, illuminate cellular signalling responses and provide a resource to link phosphorylation events to biological pathways.


Subject(s)
Phosphoproteins , Protein Serine-Threonine Kinases , Proteome , Serine , Threonine , Humans , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Serine/metabolism , Substrate Specificity , Threonine/metabolism , Proteome/chemistry , Proteome/metabolism , Datasets as Topic , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Cell Line , Phosphoserine/metabolism , Phosphothreonine/metabolism
3.
Adv Sci (Weinh) ; 9(23): e2200088, 2022 08.
Article in English | MEDLINE | ID: mdl-35607290

ABSTRACT

Reaching population immunity against COVID-19 is proving difficult even in countries with high vaccination levels. Thus, it is critical to identify limits of control and effective measures against future outbreaks. The effects of nonpharmaceutical interventions (NPIs) and vaccination strategies are analyzed with a detailed community-specific agent-based model (ABM). The authors demonstrate that the threshold for population immunity is not a unique number, but depends on the vaccination strategy. Prioritizing highly interactive people diminishes the risk for an infection wave, while prioritizing the elderly minimizes fatalities when vaccinations are low. Control over COVID-19 outbreaks requires adaptive combination of NPIs and targeted vaccination, exemplified for Germany for January-September 2021. Bimodality emerges from the heterogeneity and stochasticity of community-specific human-human interactions and infection networks, which can render the effects of limited NPIs uncertain. The authors' simulation platform can process and analyze dynamic COVID-19 epidemiological situations in diverse communities worldwide to predict pathways to population immunity even with limited vaccination.


Subject(s)
COVID-19 , Aged , COVID-19/epidemiology , COVID-19/prevention & control , Computer Simulation , Disease Outbreaks/prevention & control , Germany/epidemiology , Humans , Vaccination
4.
Cell Rep ; 34(3): 108657, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33472071

ABSTRACT

It is well known that the development of drug resistance in cancer cells can lead to changes in cell morphology. Here, we describe the use of deep neural networks to analyze this relationship, demonstrating that complex cell morphologies can encode states of signaling networks and unravel cellular mechanisms hidden to conventional approaches. We perform high-content screening of 17 cancer cell lines, generating more than 500 billion data points from ∼850 million cells. We analyze these data using a deep learning model, resulting in the identification of a continuous 27-dimension space describing all of the observed cell morphologies. From its morphology alone, we could thus predict whether a cell was resistant to ErbB-family drugs, with an accuracy of 74%, and predict the potential mechanism of resistance, subsequently validating the role of MET and insulin-like growth factor 1 receptor (IGF1R) as drivers of cetuximab resistance in in vitro models of lung and head/neck cancer.


Subject(s)
Deep Learning/standards , Drug Resistance, Neoplasm/physiology , ErbB Receptors/metabolism , Machine Learning/standards , Humans , Neural Networks, Computer , Signal Transduction
5.
Dis Model Mech ; 14(3)2021 03 01.
Article in English | MEDLINE | ID: mdl-33514588

ABSTRACT

Activating PIK3CA mutations are known 'drivers' of human cancer and developmental overgrowth syndromes. We recently demonstrated that the 'hotspot' PIK3CAH1047R variant exerts unexpected allele dose-dependent effects on stemness in human pluripotent stem cells (hPSCs). In this study, we combine high-depth transcriptomics, total proteomics and reverse-phase protein arrays to reveal potentially disease-related alterations in heterozygous cells, and to assess the contribution of activated TGFß signalling to the stemness phenotype of homozygous PIK3CAH1047R cells. We demonstrate signalling rewiring as a function of oncogenic PI3K signalling strength, and provide experimental evidence that self-sustained stemness is causally related to enhanced autocrine NODAL/TGFß signalling. A significant transcriptomic signature of TGFß pathway activation in heterozygous PIK3CAH1047R was observed but was modest and was not associated with the stemness phenotype seen in homozygous mutants. Notably, the stemness gene expression in homozygous PIK3CAH1047R hPSCs was reversed by pharmacological inhibition of NODAL/TGFß signalling, but not by pharmacological PI3Kα pathway inhibition. Altogether, this provides the first in-depth analysis of PI3K signalling in hPSCs and directly links strong PI3K activation to developmental NODAL/TGFß signalling. This work illustrates the importance of allele dosage and expression when artificial systems are used to model human genetic disease caused by activating PIK3CA mutations. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Phosphatidylinositol 3-Kinases , Pluripotent Stem Cells , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Humans , Mutation/genetics , Phosphatidylinositol 3-Kinases/metabolism , Pluripotent Stem Cells/metabolism , Signal Transduction , Transforming Growth Factor beta
6.
Curr Genomics ; 22(4): 239-243, 2021 Dec 16.
Article in English | MEDLINE | ID: mdl-35273456

ABSTRACT

According to the WHO, cancer is the second most common cause of death worldwide. The social and economic damage caused by cancer is high and rising. In Europe, the annual direct medical expenses alone amount to more than €129 billion. This results in an urgent need for new and sustainable therapeutics, which has currently not been met by the pharmaceutical industry; only 3.4% of cancer drugs entering Phase I clinical trials get to market. Phosphorylation sites are parts of the core machinery of kinase signaling networks, which are known to be dysfunctional in all types of cancer. Indeed, kinases are the second most common drug target yet. However, these inhibitors block all functions of a protein, and they commonly lead to the development of resistance and increased toxicity. To facilitate global and mechanistic modeling of cancer and clinically relevant cell signaling networks, the community will have to develop sophisticated data-driven deep-learning and mechanistic computational models that generate in silico probabilistic predictions of molecular signaling network rearrangements causally implicated in cancer.

7.
Sci Rep ; 9(1): 10865, 2019 07 26.
Article in English | MEDLINE | ID: mdl-31350469

ABSTRACT

Small molecule inhibitors of BRAF and MEK have proven effective at inhibiting tumor growth in melanoma patients, however this efficacy is limited due to the almost universal development of drug resistance. To provide advanced insight into the signaling responses that occur following kinase inhibition we have performed quantitative (phospho)-proteomics of human melanoma cells treated with either dabrafenib, a BRAF inhibitor; trametinib, a MEK inhibitor or SCH772984, an ERK inhibitor. Over nine experiments we identified 7827 class I phosphorylation sites on 4960 proteins. This included 54 phosphorylation sites that were significantly down-modulated after exposure to all three inhibitors, 34 of which have not been previously reported. Functional analysis of these novel ERK targets identified roles for them in GTPase activity and regulation, apoptosis and cell-cell adhesion. Comparison of the results presented here with previously reported phosphorylation sites downstream of ERK showed a limited degree of overlap suggesting that ERK signaling responses may be highly cell line and cue specific. In addition we identified 26 phosphorylation sites that were only responsive to dabrafenib. We provide further orthogonal experimental evidence for 3 of these sites in human embryonic kidney cells over-expressing BRAF as well as further computational insights using KinomeXplorer. The validated phosphorylation sites were found to be involved in actin regulation, which has been proposed as a novel mechanism for inhibiting resistance development. These results would suggest that the linearity of the BRAF-MEK-ERK module is at least context dependent.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Imidazoles/pharmacology , Indazoles/pharmacology , Melanoma/metabolism , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Oximes/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Pyridones/pharmacology , Pyrimidinones/pharmacology , Skin Neoplasms/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , MAP Kinase Signaling System/drug effects , Melanoma/pathology , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation/drug effects , Proteome , Proteomics/methods , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/pathology
8.
Elife ; 82019 05 24.
Article in English | MEDLINE | ID: mdl-31124786

ABSTRACT

Human NimA-related kinases (Neks) have multiple mitotic and non-mitotic functions, but few substrates are known. We systematically determined the phosphorylation-site motifs for the entire Nek kinase family, except for Nek11. While all Nek kinases strongly select for hydrophobic residues in the -3 position, the family separates into four distinct groups based on specificity for a serine versus threonine phospho-acceptor, and preference for basic or acidic residues in other positions. Unlike Nek1-Nek9, Nek10 is a dual-specificity kinase that efficiently phosphorylates itself and peptide substrates on serine and tyrosine, and its activity is enhanced by tyrosine auto-phosphorylation. Nek10 dual-specificity depends on residues in the HRD+2 and APE-4 positions that are uncommon in either serine/threonine or tyrosine kinases. Finally, we show that the phosphorylation-site motifs for the mitotic kinases Nek6, Nek7 and Nek9 are essentially identical to that of their upstream activator Plk1, suggesting that Nek6/7/9 function as phospho-motif amplifiers of Plk1 signaling.


Subject(s)
NIMA-Related Kinases/metabolism , Signal Transduction , Substrate Specificity , Humans , NIMA-Related Kinases/chemistry , Phosphorylation , Serine/metabolism , Threonine/metabolism
9.
PLoS Biol ; 17(3): e2006540, 2019 03.
Article in English | MEDLINE | ID: mdl-30897078

ABSTRACT

Specificity within protein kinase signaling cascades is determined by direct and indirect interactions between kinases and their substrates. While the impact of localization and recruitment on kinase-substrate targeting can be readily assessed, evaluating the relative importance of direct phosphorylation site interactions remains challenging. In this study, we examine the STE20 family of protein serine-threonine kinases to investigate basic mechanisms of substrate targeting. We used peptide arrays to define the phosphorylation site specificity for the majority of STE20 kinases and categorized them into four distinct groups. Using structure-guided mutagenesis, we identified key specificity-determining residues within the kinase catalytic cleft, including an unappreciated role for the kinase ß3-αC loop region in controlling specificity. Exchanging key residues between the STE20 kinases p21-activated kinase 4 (PAK4) and Mammalian sterile 20 kinase 4 (MST4) largely interconverted their phosphorylation site preferences. In cells, a reprogrammed PAK4 mutant, engineered to recognize MST substrates, failed to phosphorylate PAK4 substrates or to mediate remodeling of the actin cytoskeleton. In contrast, this mutant could rescue signaling through the Hippo pathway in cells lacking multiple MST kinases. These observations formally demonstrate the importance of catalytic site specificity for directing protein kinase signal transduction pathways. Our findings further suggest that phosphorylation site specificity is both necessary and sufficient to mediate distinct signaling outputs of STE20 kinases and imply broad applicability to other kinase signaling systems.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , p21-Activated Kinases/metabolism , Catalysis , Cell Line , Humans , Mutagenesis/genetics , Mutagenesis/physiology , Phosphorylation/genetics , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Signal Transduction/genetics , p21-Activated Kinases/genetics
10.
J Biol Chem ; 293(43): 16608-16622, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30194279

ABSTRACT

ARGONAUTE1 (AGO1) binds directly to small regulatory RNA and is a key effector protein of post-transcriptional gene silencing mediated by microRNA (miRNA) and small interfering RNA (siRNA) in Arabidopsis The formation of an RNA-induced silencing complex (RISC) of AGO1 and small RNA requires the function of the heat shock protein 70/90 chaperone system. Some functions of AGO1 occur in association with endomembranes, in particular the rough endoplasmic reticulum (RER), but proteins interacting with AGO1 in membrane fractions remain unidentified. In this study, we show that the farnesylated heat shock protein 40 homologs, J2 and J3, associate with AGO1 in membrane fractions in a manner that involves protein farnesylation. We also show that three changes in AGO1 function are detectable in mutants in protein farnesylation and J2/J3. First, perturbations of the HSP40/70/90 pathway by mutation of J3, HSP90, and farnesyl transferase affect the amounts of AGO1 associated with membranes. Second, miRNA association with membrane-bound polysomes is increased in farnesyl transferase and farnesylation-deficient J2/J3 mutants. Third, silencing by noncell autonomously acting short interfering RNAs is impaired. These observations highlight the involvement of farnesylated J2/J3 in small RNA-mediated gene regulation, and suggest that the importance of chaperone-AGO1 interaction is not limited to the RISC assembly process.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , HSP40 Heat-Shock Proteins/metabolism , RNA-Induced Silencing Complex/metabolism , Arabidopsis/genetics , Arabidopsis Proteins/genetics , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , HSP40 Heat-Shock Proteins/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Prenylation , RNA, Plant/genetics , RNA, Plant/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Induced Silencing Complex/genetics
11.
Psychiatr Genet ; 28(5): 77-84, 2018 10.
Article in English | MEDLINE | ID: mdl-30080747

ABSTRACT

OBJECTIVE: Glycogen synthase kinase 3ß (GSK3ß) has been implicated in mood disorders. We previously reported associations between a GSK3ß polymorphism and hippocampal volume in major depressive disorder (MDD). We then reported similar associations for a subset of GSK3ß-regulated genes. We now investigate an algorithm-derived comprehensive list of genes encoding proteins that directly interact with GSK3ß to identify a genotypic network influencing hippocampal volume in MDD. PARTICIPANTS AND METHODS: We used discovery (N=141) and replication (N=77) recurrent MDD samples. Our gene list was generated from the NetworKIN database. Hippocampal measures were derived using an optimized Freesurfer protocol. We identified interacting single nucleotide polymorphisms using the machine learning algorithm Random Forest and verified interactions using likelihood ratio tests between nested linear regression models. RESULTS: The discovery sample showed multiple two-single nucleotide polymorphism interactions with hippocampal volume. The replication sample showed a replicable interaction (likelihood ratio test: P=0.0088, replication sample; P=0.017, discovery sample; Stouffer's combined P=0.0007) between genes associated previously with endoplasmic reticulum stress, calcium regulation and histone modifications. CONCLUSION: Our results provide genetic evidence supporting associations between hippocampal volume and MDD, which may reflect underlying cellular stress responses. Our study provides evidence of biological mechanisms that should be further explored in the search for disease-modifying therapeutic targets for depression.


Subject(s)
Depressive Disorder, Major/genetics , Glycogen Synthase Kinase 3 beta/genetics , Hippocampus/pathology , Adult , Aged , Algorithms , Case-Control Studies , Databases, Genetic , Depressive Disorder, Major/enzymology , Depressive Disorder, Major/metabolism , Female , Gene Regulatory Networks , Genetic Predisposition to Disease , Genome-Wide Association Study , Genotype , Glycogen Synthase Kinase 3 beta/metabolism , Hippocampus/enzymology , Hippocampus/metabolism , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Polymorphism, Single Nucleotide
12.
PLoS Comput Biol ; 14(1): e1005900, 2018 01.
Article in English | MEDLINE | ID: mdl-29309407

ABSTRACT

Cell migration is a central biological process that requires fine coordination of molecular events in time and space. A deregulation of the migratory phenotype is also associated with pathological conditions including cancer where cell motility has a causal role in tumor spreading and metastasis formation. Thus cell migration is of critical and strategic importance across the complex disease spectrum as well as for the basic understanding of cell phenotype. Experimental studies of the migration of cells in monolayers are often conducted with 'wound healing' assays. Analysis of these assays has traditionally relied on how the wound area changes over time. However this method does not take into account the shape of the wound. Given the many options for creating a wound healing assay and the fact that wound shape invariably changes as cells migrate this is a significant flaw. Here we present a novel software package for analyzing concerted cell velocity in wound healing assays. Our method encompasses a wound detection algorithm based on cell confluency thresholding and employs a Bayesian approach in order to estimate concerted cell velocity with an associated likelihood. We have applied this method to study the effect of siRNA knockdown on the migration of a breast cancer cell line and demonstrate that cell velocity can track wound healing independently of wound shape and provides a more robust quantification with significantly higher signal to noise ratios than conventional analyses of wound area. The software presented here will enable other researchers in any field of cell biology to quantitatively analyze and track live cell migratory processes and is therefore expected to have a significant impact on the study of cell migration, including cancer relevant processes. Installation instructions, documentation and source code can be found at http://bowhead.lindinglab.science licensed under GPLv3.


Subject(s)
Breast Neoplasms/genetics , Cell Movement , Gene Expression Regulation, Neoplastic , Algorithms , Bayes Theorem , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Computational Biology , Female , Humans , Molecular Motor Proteins/metabolism , Myosin Heavy Chains/metabolism , Normal Distribution , Octamer Transcription Factor-3/metabolism , Phenotype , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/metabolism , Signal-To-Noise Ratio , Time Factors , Wound Healing , Polo-Like Kinase 1
14.
Cell Rep ; 20(12): 2784-2791, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28930675

ABSTRACT

Signaling networks are nonlinear and complex, involving a large ensemble of dynamic interaction states that fluctuate in space and time. However, therapeutic strategies, such as combination chemotherapy, rarely consider the timing of drug perturbations. If we are to advance drug discovery for complex diseases, it will be essential to develop methods capable of identifying dynamic cellular responses to clinically relevant perturbations. Here, we present a Bayesian dose-response framework and the screening of an oncological drug matrix, comprising 10,000 drug combinations in melanoma and pancreatic cancer cell lines, from which we predict sequentially effective drug combinations. Approximately 23% of the tested combinations showed high-confidence sequential effects (either synergistic or antagonistic), demonstrating that cellular perturbations of many drug combinations have temporal aspects, which are currently both underutilized and poorly understood.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/analysis , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Screening Assays, Antitumor , Bayes Theorem , Cell Count , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Humans , Reproducibility of Results , Time Factors
15.
Data Brief ; 5: 621-5, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26649326

ABSTRACT

The cancer secretome includes all of the macromolecules secreted by cells into their microenvironment. Cancer cell secretomes are significantly different to that of normal cells reflecting the changes that normal cells have undergone during their transition to malignancy. More importantly, cancer secretomes are known to be active mediators of both local and distant host cells and play an important role in the progression and dissemination of cancer. Here we have quantitatively profiled both the composition of breast cancer secretomes associated with osteotropism, and their modulation under normoxic and hypoxic conditions. We detect and quantify 162 secretome proteins across all conditions which show differential hypoxic induction and association with osteotropism. Mass Spectrometry proteomics data have been deposited to the ProteomeXchange Consortium with the dataset identifier PXD000397 and the complete proteomic, bioinformatic and biological analyses are reported in Cox et al. (2015) [1].

16.
Cell ; 163(1): 202-17, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26388441

ABSTRACT

Cancer cells acquire pathological phenotypes through accumulation of mutations that perturb signaling networks. However, global analysis of these events is currently limited. Here, we identify six types of network-attacking mutations (NAMs), including changes in kinase and SH2 modulation, network rewiring, and the genesis and extinction of phosphorylation sites. We developed a computational platform (ReKINect) to identify NAMs and systematically interpreted the exomes and quantitative (phospho-)proteomes of five ovarian cancer cell lines and the global cancer genome repository. We identified and experimentally validated several NAMs, including PKCγ M501I and PKD1 D665N, which encode specificity switches analogous to the appearance of kinases de novo within the kinome. We discover mutant molecular logic gates, a drift toward phospho-threonine signaling, weakening of phosphorylation motifs, and kinase-inactivating hotspots in cancer. Our method pinpoints functional NAMs, scales with the complexity of cancer genomes and cell signaling, and may enhance our capability to therapeutically target tumor-specific networks.


Subject(s)
Ovarian Neoplasms/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Signal Transduction , Female , Humans , Information Storage and Retrieval , Models, Molecular , Point Mutation , Protein Kinases/chemistry , Software
17.
Cell ; 163(1): 187-201, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26388442

ABSTRACT

Protein kinases control cellular responses to environmental cues by swift and accurate signal processing. Breakdowns in this high-fidelity capability are a driving force in cancer and other diseases. Thus, our limited understanding of which amino acids in the kinase domain encode substrate specificity, the so-called determinants of specificity (DoS), constitutes a major obstacle in cancer signaling. Here, we systematically discover several DoS and experimentally validate three of them, named the αC1, αC3, and APE-7 residues. We demonstrate that DoS form sparse networks of non-conserved residues spanning distant regions. Our results reveal a likely role for inter-residue allostery in specificity and an evolutionary decoupling of kinase activity and specificity, which appear loaded on independent groups of residues. Finally, we uncover similar properties driving SH2 domain specificity and demonstrate how the identification of DoS can be utilized to elucidate a greater understanding of the role of signaling networks in cancer (Creixell et al., 2015 [this issue of Cell]).


Subject(s)
Protein Kinases/chemistry , Protein Kinases/metabolism , Computational Biology , Humans , Models, Molecular , Neoplasms/metabolism , Substrate Specificity , src Homology Domains
18.
Nat Methods ; 12(7): 615-621, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26125594

ABSTRACT

Genomic information on tumors from 50 cancer types cataloged by the International Cancer Genome Consortium (ICGC) shows that only a few well-studied driver genes are frequently mutated, in contrast to many infrequently mutated genes that may also contribute to tumor biology. Hence there has been large interest in developing pathway and network analysis methods that group genes and illuminate the processes involved. We provide an overview of these analysis techniques and show where they guide mechanistic and translational investigations.


Subject(s)
Gene Regulatory Networks , Genome , Neoplasms/genetics , Signal Transduction/physiology , Humans
19.
Nature ; 522(7554): 106-110, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26017313

ABSTRACT

Tumour metastasis is a complex process involving reciprocal interplay between cancer cells and host stroma at both primary and secondary sites, and is strongly influenced by microenvironmental factors such as hypoxia. Tumour-secreted proteins play a crucial role in these interactions and present strategic therapeutic potential. Metastasis of breast cancer to the bone affects approximately 85% of patients with advanced disease and renders them largely untreatable. Specifically, osteolytic bone lesions, where bone is destroyed, lead to debilitating skeletal complications and increased patient morbidity and mortality. The molecular interactions governing the early events of osteolytic lesion formation are currently unclear. Here we show hypoxia to be specifically associated with bone relapse in patients with oestrogen-receptor negative breast cancer. Global quantitative analysis of the hypoxic secretome identified lysyl oxidase (LOX) as significantly associated with bone-tropism and relapse. High expression of LOX in primary breast tumours or systemic delivery of LOX leads to osteolytic lesion formation whereas silencing or inhibition of LOX activity abrogates tumour-driven osteolytic lesion formation. We identify LOX as a novel regulator of NFATc1-driven osteoclastogenesis, independent of RANK ligand, which disrupts normal bone homeostasis leading to the formation of focal pre-metastatic lesions. We show that these lesions subsequently provide a platform for circulating tumour cells to colonize and form bone metastases. Our study identifies a novel mechanism of regulation of bone homeostasis and metastasis, opening up opportunities for novel therapeutic intervention with important clinical implications.


Subject(s)
Bone Neoplasms/pathology , Bone Neoplasms/secondary , Breast Neoplasms/enzymology , Breast Neoplasms/metabolism , Neoplasm Metastasis , Protein-Lysine 6-Oxidase/metabolism , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/prevention & control , Breast Neoplasms/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Female , Humans , Mice , NFATC Transcription Factors/metabolism , Neoplasm Metastasis/pathology , Neoplastic Cells, Circulating/pathology , Osteoblasts/metabolism , Osteoblasts/pathology , Osteoclasts/metabolism , Osteoclasts/pathology , Protein-Lysine 6-Oxidase/antagonists & inhibitors , Receptors, Estrogen/deficiency , Receptors, Estrogen/genetics
20.
Sci Signal ; 8(371): rs3, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25852190

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is an endogenous secreted peptide and, in preclinical studies, preferentially induces apoptosis in tumor cells rather than in normal cells. The acquisition of resistance in cells exposed to TRAIL or its mimics limits their clinical efficacy. Because kinases are intimately involved in the regulation of apoptosis, we systematically characterized kinases involved in TRAIL signaling. Using RNA interference (RNAi) loss-of-function and cDNA overexpression screens, we identified 169 protein kinases that influenced the dynamics of TRAIL-induced apoptosis in the colon adenocarcinoma cell line DLD-1. We classified the kinases as sensitizers or resistors or modulators, depending on the effect that knockdown and overexpression had on TRAIL-induced apoptosis. Two of these kinases that were classified as resistors were PX domain-containing serine/threonine kinase (PXK) and AP2-associated kinase 1 (AAK1), which promote receptor endocytosis and may enable cells to resist TRAIL-induced apoptosis by enhancing endocytosis of the TRAIL receptors. We assembled protein interaction maps using mass spectrometry-based protein interaction analysis and quantitative phosphoproteomics. With these protein interaction maps, we modeled information flow through the networks and identified apoptosis-modifying kinases that are highly connected to regulated substrates downstream of TRAIL. The results of this analysis provide a resource of potential targets for the development of TRAIL combination therapies to selectively kill cancer cells.


Subject(s)
Adenocarcinoma/metabolism , Apoptosis , Colonic Neoplasms/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Protein Serine-Threonine Kinases/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...