Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 14(23)2022 Nov 22.
Article in English | MEDLINE | ID: mdl-36497220

ABSTRACT

BACKGROUND: Cases of the spontaneous regression of multiple pulmonary metastases, after radiofrequency ablation (RFA), of a single lung metastasis, have been documented to be mediated by the immune system. The interaction of immune checkpoints, e.g., PD-1/PD-L1 and CTLA-4/CD80, may explain this phenomenon. The purpose of this study is to identify and quantify immune mechanisms triggered by RFA of pulmonary metastases originating from colorectal cancer. METHODS: We used two-site time-resolved Förster resonance energy transfer as determined by frequency-domain FLIM (iFRET) for the quantification of receptor-ligand interactions. iFRET provides a method by which immune checkpoint interaction states can be quantified in a spatiotemporal manner. The same patient sections were used for assessment of ligand-receptor interaction and intratumoral T-cell labeling. CONCLUSION: The checkpoint interaction states quantified by iFRET did not correlate with ligand expression. We show that immune checkpoint ligand expression as a predictive biomarker may be unsuitable as it does not confirm checkpoint interactions. In pre-RFA-treated metastases, there was a significant and negative correlation between PD-1/PD-L1 interaction state and intratumoral CD3+ and CD8+ density. The negative correlation of CD8+ and interactive states of PD-1/PD-L1 can be used to assess the state of immune suppression in RFA-treated patients.

2.
FEBS Lett ; 596(21): 2721-2735, 2022 11.
Article in English | MEDLINE | ID: mdl-36002439

ABSTRACT

Innate and adaptive immune systems are built-in homeostatic functions of many multicellular organisms and protect the host against foreign pathogens and infections. Dysregulation of the molecular mechanisms of the immune system can result in autoimmune diseases. The immune system can also be harnessed and manipulated to provide targeted cancer therapies, some of them relying on the blockade of immune-checkpoint receptors. Two prominent immune checkpoints, PD-1/PD-L1 and CTLA-4/CD80, comprise receptor-ligand pairs that prevent the host immune cells from attacking host tissues. However, cancer cells upregulate the respective PD-L1 and CD80 ligands for PD-1 and CTLA-4 and thereby evade the host-immune response. Therapeutic drugs that block PD-1/PD-L1 and CTLA-4/CD80 interactions re-enable the immune system to attack cancer cells, but their prognostic biomarker remains challenging. In this review, we discuss how the use of quantitative molecular imaging can be exploited to predict the response to anti-PD-1/PD-L1 therapies and to identify cancer patients who would benefit from them.


Subject(s)
B7-H1 Antigen , Immunotherapy , Neoplasms , Precision Medicine , Programmed Cell Death 1 Receptor , Proteomics , Humans , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Immunotherapy/methods , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/immunology , Precision Medicine/methods , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Proteomics/methods
3.
Biophys Chem ; 283: 106768, 2022 04.
Article in English | MEDLINE | ID: mdl-35114593

ABSTRACT

Oncogene dysregulation is a driver of neoplasia development and progression. The use of quantitative molecular imaging to quantify oncogene activation will be crucial in developing companion diagnostics which can identify personalised patient regimens. However, the evaluation of oncogene activation does not necessarily correlate with oncoprotein activation. Post-translational modifications, such as phosphorylation, lipidation and methylation, may enhance oncoprotein functionality. It is this functionality that progresses neoplasia and may correlate with patient outcome. Advanced molecular imaging may be used to directly quantify oncoprotein, as opposed to oncogene, activation. Time-resolved Förster Resonance Energy Transfer (TR-FRET) involves the non-radiative transfer of energy from one chromophore to another over distances of 1-10 nm; allowing FRET to be used as a "chemical ruler". TR-FRET can be utilised to directly elucidate spatial oncoprotein activation in single cells and patient tissues. In single cells, TR-FRET has uncovered the mechanisms by which PKCß1 is trafficked to the nucleus and cleaved. Additionally it has revealed the mechanism of activation of Akt/PKB, whereby Akt/PKB undergoes a conformational change, allowing the Thr308 site to be phosphorylated by PDK1. Moreover TR-FRET has been utilised to quantify HER2-HER heterodimerisation and Akt/PKB activation states in patient biopsies, where it is shown to be predictive of outcome/relapse. The role of TR-FRET is not solely limited to intracellular signalling events. A study has used TR-FRET to measure intercellular immune-checkpoint receptor-ligand interactions. Within this study it was seen that PD-L1 expression was not indicative of PD-1/PD-L1 interaction states in a range of solid tumours. Crucially, in melanoma and NSCLC, PD-1/PD-L1 interaction was a predictive of an improved patient outcome. PD-L1 expression did not predict patient outcome. Several groups have worked to improve Fluorescence lifetime imaging microscopy (FLIM) acquisition times, including the use of: window-galvanometers; multifocal multiphoton FLIM and parallel pixel excitation coupled with wide-field time-gated FLIM. The development of novel quantitative molecular imaging will be critical in the development of personalised patient therapies in the future.


Subject(s)
B7-H1 Antigen , Proto-Oncogene Proteins c-akt , Fluorescence Resonance Energy Transfer/methods , Humans , Microscopy, Fluorescence/methods , Programmed Cell Death 1 Receptor
4.
Biochim Biophys Acta Biomembr ; 1864(1): 183780, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34547252

ABSTRACT

The general segregation of a molecular class, lipids, from the pathways of cellular communication, via endo-membranes, has resulted in the over-simplification and misconceptions in deciphering cell signalling mechanisms. Mechanisms in signal transduction and protein activation require targeting of proteins to membranous compartments with a specific localised morphology and dynamics that are dependent on their lipid composition. Many posttranslational events define cellular behaviours and without the active role of membranous compartments these events lead to various dysregulations of the signalling pathways. We summarise the key findings, using tools such as the rapalogue dimerisation, in the structural roles and signalling of the inter-related phosphoinositide lipids and their derivative, diacylglycerol, in the regulation of nuclear envelope biogenesis and other subcellular compartments such as the nucleoplasmic reticulum.


Subject(s)
Cell Communication/genetics , Membrane Lipids/genetics , Phosphatidylinositols/genetics , Signal Transduction/genetics , Nuclear Envelope/genetics
5.
Gastroenterology ; 161(4): 1179-1193, 2021 10.
Article in English | MEDLINE | ID: mdl-34197832

ABSTRACT

BACKGROUND & AIMS: Colorectal cancer (CRC) shows variable response to immune checkpoint blockade, which can only partially be explained by high tumor mutational burden (TMB). We conducted an integrated study of the cancer tissue and associated tumor microenvironment (TME) from patients treated with pembrolizumab (KEYNOTE 177 clinical trial) or nivolumab to dissect the cellular and molecular determinants of response to anti- programmed cell death 1 (PD1) immunotherapy. METHODS: We selected multiple regions per tumor showing variable T-cell infiltration for a total of 738 regions from 29 patients, divided into discovery and validation cohorts. We performed multiregional whole-exome and RNA sequencing of the tumor cells and integrated these with T-cell receptor sequencing, high-dimensional imaging mass cytometry, detection of programmed death-ligand 1 (PDL1) interaction in situ, multiplexed immunofluorescence, and computational spatial analysis of the TME. RESULTS: In hypermutated CRCs, response to anti-PD1 immunotherapy was not associated with TMB but with high clonality of immunogenic mutations, clonally expanded T cells, low activation of Wnt signaling, deregulation of the interferon gamma pathway, and active immune escape mechanisms. Responsive hypermutated CRCs were also rich in cytotoxic and proliferating PD1+CD8 T cells interacting with PDL1+ antigen-presenting macrophages. CONCLUSIONS: Our study clarified the limits of TMB as a predictor of response of CRC to anti-PD1 immunotherapy. It identified a population of antigen-presenting macrophages interacting with CD8 T cells that consistently segregate with response. We therefore concluded that anti-PD1 agents release the PD1-PDL1 interaction between CD8 T cells and macrophages to promote cytotoxic antitumor activity.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Colorectal Neoplasms/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Immunogenetic Phenomena , Immunogenetics , Nivolumab/therapeutic use , Tumor Microenvironment , Antibodies, Monoclonal, Humanized/adverse effects , Biomarkers, Tumor/genetics , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Clinical Trials as Topic , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Cytotoxicity, Immunologic/drug effects , Gene Expression Profiling , Humans , Immune Checkpoint Inhibitors/adverse effects , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mutation , Nivolumab/adverse effects , Predictive Value of Tests , Programmed Cell Death 1 Receptor/antagonists & inhibitors , RNA-Seq , Reproducibility of Results , Time Factors , Transcriptome , Treatment Outcome , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology , Exome Sequencing
7.
Cancer Res ; 80(19): 4244-4257, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32855204

ABSTRACT

Many cancers are termed immunoevasive due to expression of immunomodulatory ligands. Programmed death ligand-1 (PD-L1) and cluster of differentiation 80/86 (CD80/86) interact with their receptors, programmed death receptor-1 (PD-1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4), respectively, on tumor-infiltrating leukocytes eliciting immunosuppression. Immunotherapies aimed at blocking these interactions are revolutionizing cancer treatments, albeit in an inadequately described patient subset. To address the issue of patient stratification for immune checkpoint intervention, we quantitatively imaged PD-1/PD-L1 interactions in tumor samples from patients, employing an assay that readily detects these intercellular protein-protein interactions in the less than or equal to 10 nm range. These analyses across multiple patient cohorts demonstrated the intercancer, interpatient, and intratumoral heterogeneity of interacting immune checkpoints. The PD-1/PD-L1 interaction was not correlated with clinical PD-L1 expression scores in malignant melanoma. Crucially, among anti-PD-1-treated patients with metastatic non-small cell lung cancer, those with lower PD-1/PD-L1 interaction had significantly worsened survival. It is surmised that within tumors selecting for an elevated level of PD-1/PD-L1 interaction, there is a greater dependence on this pathway for immune evasion and hence, they exhibit more impressive patient response to intervention. SIGNIFICANCE: Quantitation of immune checkpoint interaction by direct imaging demonstrates that immunotherapy-treated patients with metastatic NSCLC with a low extent of PD-1/PD-L1 interaction show significantly worse outcome.


Subject(s)
B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Renal Cell/immunology , Kidney Neoplasms/immunology , Lung Neoplasms/immunology , Melanoma/immunology , Programmed Cell Death 1 Receptor/metabolism , Adult , Aged , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Female , Fluorescence Resonance Energy Transfer/methods , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/mortality , Middle Aged , Molecular Targeted Therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Reproducibility of Results , Treatment Outcome
8.
Anal Chem ; 92(10): 6858-6868, 2020 05 19.
Article in English | MEDLINE | ID: mdl-32324380

ABSTRACT

The human nuclear membrane is composed of a double bilayer, the inner membrane being linked to the protein lamina network and the outer nuclear membrane continuous with the endoplasmic reticulum. Nuclear membranes can form large invaginations inside the nucleus; their specific roles still remain unknown. Although much of the protein identification has been determined, their lipid composition remains largely undetermined. In order to understand the mechanical and dynamic properties of nuclear membranes we investigated their lipid composition by two quantitative methods, namely, 31P and 1H multidimensional NMR and mass spectrometry, using internal standards. We also developed a nondetergent nuclei extraction protocol allowing to produce milligram quantities of nuclear membrane lipids. We found that the nuclear membrane lipid extract is composed of a complex mixture of phospholipids with different phosphatidylcholine species present in large amounts. Negatively charged lipids, with elevated amounts of phosphatidylinositol (PI), were also present. Mass spectrometry confirmed the phospholipid composition and provided further information on acyl-chain length and unsaturation. Lipid chain lengths ranged between 30 and 38 carbon atoms (two chains summed up) with a high proportion of 34 carbon atom length for most species. PI lipids have high amounts of chain lengths with 36-38 carbons. Independent of the chain length unsaturations were highly elevated with one to two double bonds per lipid species.


Subject(s)
Cell Nucleus/chemistry , Membrane Lipids/analysis , HEK293 Cells , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry
9.
Sci Rep ; 10(1): 5147, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32198481

ABSTRACT

Human nuclear membrane (hNM) invaginations are thought to be crucial in fusion, fission and remodeling of cells and present in many human diseases. There is however little knowledge, if any, about their lipid composition and dynamics. We therefore isolated nuclear envelope lipids from human kidney cells, analyzed their composition and determined the membrane dynamics after resuspension in buffer. The hNM lipid extract was composed of a complex mixture of phospholipids, with high amounts of phosphatidylcholines, phosphatidylinositols (PI) and cholesterol. hNM dynamics was determined by solid-state NMR and revealed that the lamellar gel-to-fluid phase transition occurs below 0 °C, reflecting the presence of elevated amounts of unsaturated fatty acid chains. Fluidity was higher than the plasma membrane, illustrating the dual action of Cholesterol (ordering) and PI lipids (disordering). The most striking result was the large magnetic field-induced membrane deformation allowing to determine the membrane bending elasticity, a property related to hydrodynamics of cells and organelles. Human Nuclear Lipid Membranes were at least two orders of magnitude more elastic than the classical plasma membrane suggesting a physical explanation for the formation of nuclear membrane invaginations.


Subject(s)
Membrane Fluidity/physiology , Nuclear Envelope/metabolism , Nuclear Envelope/physiology , Cell Membrane/metabolism , Cholesterol/metabolism , Fatty Acids, Unsaturated/metabolism , Humans , Kidney/pathology , Magnetic Fields , Magnetic Resonance Spectroscopy , Membrane Lipids/metabolism , Phase Transition , Phosphatidylinositols/metabolism , Phospholipids/metabolism
10.
Sci Rep ; 9(1): 14527, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31601855

ABSTRACT

3'-Phosphoinositide-dependent-Kinase-1 (PDK1) is a master regulator whereby its PI3-kinase-dependent dysregulation in human pathologies is well documented. Understanding the direct role for PtdIns(3,4,5)P3 and other anionic phospholipids in the regulation of PDK1 conformational dynamics and its downstream activation remains incomplete. Using advanced quantitative-time-resolved imaging (Fluorescence Lifetime Imaging and Fluorescence Correlation Spectroscopy) and molecular modelling, we show an interplay of antagonistic binding effects of PtdIns(3,4,5)P3 and other anionic phospholipids, regulating activated PDK1 homodimers. We demonstrate that phosphatidylserine maintains PDK1 in an inactive conformation. The dysregulation of the PI3K pathway affects the spatio-temporal and conformational dynamics of PDK1 and the activation of its downstream substrates. We have established a new anionic-phospholipid-dependent model for PDK1 regulation, depicting the conformational dynamics of multiple homodimer states. We show that the dysregulation of the PI3K pathway perturbs equilibrium between the PDK1 homodimer conformations. Our findings provide a role for the PtdSer binding site and its previously unrewarding role in PDK1 downregulation, suggesting a possible therapeutic strategy where the constitutively active dimer conformer of PDK1 may be rendered inactive by small molecules that drive it to its PtdSer-bound conformer.


Subject(s)
Anions/chemistry , Phospholipids/chemistry , Protein Multimerization , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/chemistry , Animals , Green Fluorescent Proteins/chemistry , Humans , Lipids/chemistry , Mice , Microscopy, Confocal , Models, Molecular , NIH 3T3 Cells , Phosphatidylinositol 3-Kinases/chemistry , Phosphatidylinositol Phosphates , Phosphorylation , Protein Binding , Protein Conformation , Recombinant Proteins/chemistry
11.
J Lipid Res ; 60(11): 1880-1891, 2019 11.
Article in English | MEDLINE | ID: mdl-31548365

ABSTRACT

The ER is a large multifunctional organelle of eukaryotic cells. Malfunction of the ER in various disease states, such as atherosclerosis, diabetes, cancer, Alzheimer's and Parkinson's and amyotrophic lateral sclerosis, often correlates with alterations in its morphology. The ER exhibits regionally variable membrane morphology that includes, at the extremes, large relatively flat surfaces and interconnected tubular structures highly curved in cross-section. ER morphology is controlled by shaping proteins that associate with membrane lipids. To investigate the role of these lipids, we developed a sea urchin oocyte model, a relatively quiescent cell in which the ER consists mostly of tubules. We altered levels of endogenous diacylglycerol (DAG), phosphatidylethanolamine (PtdEth), and phosphatidylcholine by microinjection of enzymes or lipid delivery by liposomes and evaluated shape changes with 2D and 3D confocal imaging and 3D electron microscopy. Decreases and increases in the levels of lipids such as DAG or PtdEth characterized by negative spontaneous curvature correlated with conversion to sheet structures or tubules, respectively. The effects of endogenous alterations of DAG were reversible upon exogenous delivery of lipids of negative spontaneous curvature. These data suggest that proteins require threshold amounts of such lipids and that localized deficiencies of the lipids could contribute to alterations of ER morphology. The oocyte modeling system should be beneficial to studies directed at understanding requirements of lipid species in interactions leading to alterations of organelle shaping.


Subject(s)
Endoplasmic Reticulum/metabolism , Oocytes/cytology , Phospholipids/metabolism , Sea Urchins , Animals , Protein Biosynthesis
12.
J Lipid Res ; 59(8): 1402-1413, 2018 08.
Article in English | MEDLINE | ID: mdl-29895700

ABSTRACT

Dysregulation of nuclear envelope (NE) assembly results in various cancers; for example, renal and some lung carcinomas ensue due to NE malformation. The NE is a dynamic membrane compartment and its completion during mitosis is a highly regulated process, but the detailed mechanism still remains incompletely understood. Previous studies have found that isolated diacylglycerol (DAG)-containing vesicles are essential for completing the fusion of the NE in nonsomatic cells. We investigated the impact of DAG depletion from the cis-Golgi in mammalian cells on NE reassembly. Using advanced electron microscopy, we observed an enriched DAG population of vesicles at the vicinity of the NE gaps of telophase mammalian cells. We applied a mini singlet oxygen generator-C1-domain tag that localized DAG-enriched vesicles at the perinuclear region, which suggested the existence of NE fusogenic vesicles. We quantified the impact of Golgi-DAG depletion by measuring the in situ NE rim curvature of the reforming NE. The rim curvature in these cells was significantly reduced compared with controls, which indicated a localized defect in NE morphology. Our novel results demonstrate the significance of the role of DAG from the cis-Golgi for the regulation of NE assembly.


Subject(s)
Diglycerides/metabolism , Golgi Apparatus/metabolism , Mitosis , Nuclear Envelope/metabolism , HeLa Cells , Humans
13.
J Biophotonics ; 11(1)2018 01.
Article in English | MEDLINE | ID: mdl-28485056

ABSTRACT

Quantification of the intracellular equilibrium dissociation constant of the interaction, Kd , is challenging due to the variability of the relative concentrations of the interacting proteins in the cell. Fluorescence lifetime imaging microscopy (FLIM) of the donor provides an accurate measurement of the molecular fraction of donor involved in FRET, but the fraction of bound acceptor is also needed to reliably estimate Kd . We present a method that exploits the spectroscopic properties of the widely used eGFP - mCherry FRET pair to rigorously determine the intracellular Kd based on imaging the fluorescence lifetime of only the donor (single-channel FLIM). We have assessed the effect of incomplete labelling and determined its range of application for different Kd using Monte Carlo simulations. We have demonstrated this method estimating the intracellular Kd for the homodimerisaton of the oncogenic protein 3-phosphoinositide-dependent kinase 1 (PDK1) in different cell lines and conditions, revealing a competitive mechanism for its regulation. The measured intracellular Kd was validated against in-vitro data. This method provides an accurate and generic tool to quantify protein interactions in situ.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Intracellular Space/metabolism , Cell Line, Tumor , Humans , Microscopy, Fluorescence , Protein Multimerization , Protein Serine-Threonine Kinases/chemistry , Protein Structure, Quaternary , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Time Factors
14.
Oncotarget ; 8(49): 84685-84696, 2017 Oct 17.
Article in English | MEDLINE | ID: mdl-29156676

ABSTRACT

Mammalian target of rapamycin (mTOR) is a central regulator of growth and metabolism. mTOR resides in two distinct multi-protein complexes - mTORC1 and mTORC2 - with distinct upstream regulators and downstream targets. While it is possible to specifically inhibit mTORC1 with rapamycin, or inhibit both mTOR complexes together with ATP pocket directed mTOR kinase inhibitors, it is not possible to assess the specific roles for mTORC2 pharmacologically. To overcome this, we have developed a novel, inducible, dominant negative system for disrupting substrate recruitment to mTORC2. Previously we identified the mTORC2 specific subunit Sin1 as a direct binding partner for AGC kinases Akt and PKC. Sin1 mutants, which retain the ability to bind Rictor and mTOR, but fail to recruit their AGC client kinases, inhibit AKT and PKC priming and block cell growth. In this study, we demonstrate that uncoupling mTORC2 from AGC kinases in DLD1 colon cancer cells inhibits Akt activation and blocks tumour growth in vivo. Further we demonstrate, using time resolved two-site amplified FRET (A-FRET) analysis of xenograft tumours, that inhibition of tumour growth correlates with the degree of mTORC2 uncoupling from its downstream targets, as demonstrated for Akt. These data add weight to the body of evidence that mTORC2 represents a pharmacological target in cancer independently of mTORC1.

15.
J Chem Biol ; 10(4): 157, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29075353
16.
J Struct Biol ; 199(2): 120-131, 2017 08.
Article in English | MEDLINE | ID: mdl-28576556

ABSTRACT

Super-resolution light microscopy, correlative light and electron microscopy, and volume electron microscopy are revolutionising the way in which biological samples are examined and understood. Here, we combine these approaches to deliver super-accurate correlation of fluorescent proteins to cellular structures. We show that YFP and GFP have enhanced blinking properties when embedded in acrylic resin and imaged under partial vacuum, enabling in vacuo single molecule localisation microscopy. In conventional section-based correlative microscopy experiments, the specimen must be moved between imaging systems and/or further manipulated for optimal viewing. These steps can introduce undesirable alterations in the specimen, and complicate correlation between imaging modalities. We avoided these issues by using a scanning electron microscope with integrated optical microscope to acquire both localisation and electron microscopy images, which could then be precisely correlated. Collecting data from ultrathin sections also improved the axial resolution and signal-to-noise ratio of the raw localisation microscopy data. Expanding data collection across an array of sections will allow 3-dimensional correlation over unprecedented volumes. The performance of this technique is demonstrated on vaccinia virus (with YFP) and diacylglycerol in cellular membranes (with GFP).


Subject(s)
Luminescent Proteins/analysis , Microscopy, Electron, Scanning/methods , Microscopy, Fluorescence/methods , Single Molecule Imaging/methods , Bacterial Proteins/analysis , Diglycerides/analysis , Equipment Design , Green Fluorescent Proteins/analysis , Signal-To-Noise Ratio , Vacuum
18.
BBA Clin ; 8: 97-102, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29296589

ABSTRACT

PURPOSE: Clear cell Renal Cell Carcinomas (ccRCC), the largest group of renal tumours, are resistant to classical therapies. The determination of the functional state of actionable biomarkers for the assessment of these adenocarcinomas is essential. The dysregulation of the oncoprotein, PKB/Akt has been linked with poor prognoses in human cancers. MATERIAL & METHODS: We analysed the status of the PKB/Akt pathway in a representative tumour tissue microarray obtained from the primary tumours and their metastases in 60 ccRCC with long term follow up. We sought to define the evolution of this pathway from the primary tumour to the metastatic event and to know the impact of its functional state in tumour aggressiveness and patient survival. Two-site time resolved amplified FRET (A-FRET) was utilised for assessing the activation state of PKB/Akt and this was compared to conventional immunohistochemistry measurements. RESULTS: Activation state of PKB/Akt in primary tumours defined by A-FRET correlated with poorer overall survival (hazard ratio 0.228; p = 0.002). Whereas, increased protein expression of phosphoPKB/Akt, identified using classical immunohistochemistry, yielded no significant difference (hazard ratio 1.390; p = 0.548). CONCLUSIONS: Quantitative determination of PKB/Akt activation in ccRCC primary tumours alongside other diagnostics tools could prove key in taking oncologists closer to an efficient personalised therapy in ccRCC patients. GENERAL SIGNIFICANCE: The quantitative imaging technology based on Amplified-FRET can rapidly analyse protein activation states and molecular interactions. It could be used for prognosis and assess drug function during the early cycles of chemotherapy. It enables evaluation of clinical efficiency of personalised cancer treatment.

19.
J Cell Sci ; 130(2): 444-452, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27927752

ABSTRACT

Regulation of nuclear envelope dynamics is an important example of the universal phenomena of membrane fusion. The signalling molecules involved in nuclear membrane fusion might also be conserved during the formation of both pronuclear and zygote nuclear envelopes in the fertilised egg. Here, we determine that class-I phosphoinositide 3-kinases (PI3Ks) are needed for in vitro nuclear envelope formation. We show that, in vivo, PtdIns(3,4,5)P3 is transiently located in vesicles around the male pronucleus at the time of nuclear envelope formation, and around male and female pronuclei before membrane fusion. We illustrate that class-I PI3K activity is also necessary for fusion of the female and male pronuclear membranes. We demonstrate, using coincidence amplified Förster resonance energy transfer (FRET) monitored using fluorescence lifetime imaging microscopy (FLIM), a protein-lipid interaction of Rab7 GTPase and PtdIns(3,4,5)P3 that occurs during pronuclear membrane fusion to create the zygote nuclear envelope. We present a working model, which includes several molecular steps in the pathways controlling fusion of nuclear envelope membranes.


Subject(s)
Membrane Fusion , Nuclear Envelope/metabolism , Paracentrotus/metabolism , Phosphatidylinositol Phosphates/metabolism , Transport Vesicles/metabolism , Zygote/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Female , Fertilization , Male , Models, Biological , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , rab7 GTP-Binding Proteins
20.
Sci Rep ; 6: 26142, 2016 05 20.
Article in English | MEDLINE | ID: mdl-27199173

ABSTRACT

Strong evidence suggests that phospholipase Cγ1 (PLCγ1) is a suitable target to counteract tumourigenesis and metastasis dissemination. We recently identified a novel signalling pathway required for PLCγ1 activation which involves formation of a protein complex with 3-phosphoinositide-dependent protein kinase 1 (PDK1). In an effort to define novel strategies to inhibit PLCγ1-dependent signals we tested here whether a newly identified and highly specific PDK1 inhibitor, 2-O-benzyl-myo-inositol 1,3,4,5,6-pentakisphosphate (2-O-Bn-InsP5), could affect PDK1/PLCγ1 interaction and impair PLCγ1-dependent cellular functions in cancer cells. Here, we demonstrate that 2-O-Bn-InsP5 interacts specifically with the pleckstrin homology domain of PDK1 and impairs formation of a PDK1/PLCγ1 complex. 2-O-Bn-InsP5 is able to inhibit the epidermal growth factor-induced PLCγ1 phosphorylation and activity, ultimately resulting in impaired cancer cell migration and invasion. Importantly, we report that 2-O-Bn-InsP5 inhibits cancer cell dissemination in zebrafish xenotransplants. This work demonstrates that the PDK1/PLCγ1 complex is a potential therapeutic target to prevent metastasis and it identifies 2-O-Bn-InsP5 as a leading compound for development of anti-metastatic drugs.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Inositol Phosphates/pharmacology , Phospholipase C gamma/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Breast Neoplasms/drug therapy , Cell Line , Cell Movement/drug effects , Disease Models, Animal , Heterografts , Humans , Melanoma/drug therapy , Neoplasm Transplantation , Protein Binding , Protein Multimerization , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL