Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 168
Filter
1.
Endocrinol Diabetes Metab ; 7(2): e00475, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38475903

ABSTRACT

BACKGROUND: Sodium glucose-linked transporter 2 (SGLT2) inhibitors promote glucose, and therefore calorie, excretion in the urine. Patients taking SGLT2 inhibitors typically experience mild weight loss, but the amount of weight loss falls short of what is expected based on caloric loss. Understanding the mechanisms responsible for this weight loss discrepancy is imperative, as strategies to improve weight loss could markedly improve type 2 diabetes management and overall metabolic health. METHODS: Two mouse models of diet-induced obesity were administered the SGLT2 inhibitor empagliflozin in the food for 3 months. Urine glucose excretion, body weight, food intake and activity levels were monitored. In addition, serum hormone measurements were taken, and gene expression analyses were conducted. RESULTS: In both mouse models, mice receiving empagliflozin gained the same amount of body weight as their diet-matched controls despite marked glucose loss in the urine. No changes in food intake, serum ghrelin concentrations or activity levels were observed, but serum levels of fibroblast growth factor 21 (FGF21) decreased after treatment. A decrease in the levels of deiodinase 2 (Dio2) was also observed in the white adipose tissue, a primary target tissue of FGF21. CONCLUSION: These findings suggest that compensatory metabolic adaptations, other than increased food intake or decreased physical activity, occur in response to SGLT2 inhibitor-induced glycosuria that combats weight loss, and that reductions in FGF21, along with subsequent reductions in peripheral Dio2, may play a role.


Subject(s)
Benzhydryl Compounds , Diabetes Mellitus, Type 2 , Glucosides , Glycosuria , Sodium-Glucose Transporter 2 Inhibitors , Humans , Mice , Animals , Diet, High-Fat , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Glycosuria/metabolism , Glucose/metabolism , Body Weight , Weight Gain , Weight Loss , Eating
2.
bioRxiv ; 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38405732

ABSTRACT

The PEAK family of pseudokinases, comprising PEAK1-3, are signalling scaffolds that play oncogenic roles in several poor prognosis human cancers, including triple negative breast cancer (TNBC). However, therapeutic targeting of pseudokinases is challenging due to their lack of catalytic activity. To address this, we screened for PEAK1 effectors by affinity purification and mass spectrometry, identifying calcium/calmodulin-dependent protein kinase 2 (CAMK2)D and CAMK2G. PEAK1 promoted CAMK2D/G activation in TNBC cells via a novel feed-forward mechanism involving PEAK1/PLCγ1/Ca 2+ signalling and direct binding via a consensus CAMK2 interaction motif in the PEAK1 N-terminus. In turn, CAMK2 phosphorylated PEAK1 to enhance association with PEAK2, which is critical for PEAK1 oncogenic signalling. To achieve pharmacologic targeting of PEAK1/CAMK2, we repurposed RA306, a second generation CAMK2 inhibitor under pre-clinical development for treatment of cardiovascular disease. RA306 demonstrated on-target activity against CAMK2 in TNBC cells and inhibited PEAK1-enhanced migration and invasion in vitro . Moreover, RA306 significantly attenuated TNBC xenograft growth and blocked metastasis in a manner mirrored by CRISPR-mediated PEAK1 ablation. Overall, these studies establish PEAK1 as a critical cell signalling nexus, identify a novel mechanism for regulation of Ca 2+ signalling and its integration with tyrosine kinase signals, and identify CAMK2 as a therapeutically 'actionable' target downstream of PEAK1.

3.
iScience ; 27(3): 109031, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38380257

ABSTRACT

The transcriptional co-activator YAP forms complexes with distinct transcription factors, controlling cell fate decisions, such as proliferation and apoptosis. However, the mechanisms underlying its context-dependent function are poorly defined. This study explores the interplay between the TGF-ß and Hippo pathways and their influence on YAP's association with specific transcription factors. By integrating iterative mathematical modeling with experimental validation, we uncover molecular switches, predominantly controlled by RASSF1A and ITCH, which dictate the formation of YAP-SMAD (proliferative) and YAP-p73 (apoptotic) complexes. Our results show that RASSF1A enhances the formation of apoptotic complexes, whereas ITCH promotes the formation of proliferative complexes. Notably, higher levels of ITCH transform YAP-SMAD activity from a transient to a sustained state, impacting cellular behaviors. Extending these findings to various breast cancer cell lines highlights the role of cellular context in YAP regulation. Our study provides new insights into the mechanisms of YAP transcriptional activities and their therapeutic implications.

4.
NPJ Precis Oncol ; 8(1): 20, 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38273040

ABSTRACT

Utility of PI3Kα inhibitors like BYL719 is limited by the acquisition of genetic and non-genetic mechanisms of resistance which cause disease recurrence. Several combination therapies based on PI3K inhibition have been proposed, but a way to systematically prioritize them for breast cancer treatment is still missing. By integrating published and in-house studies, we have developed in silico models that quantitatively capture dynamics of PI3K signaling at the network-level under a BYL719-sensitive versus BYL719 resistant-cell state. Computational predictions show that signal rewiring to alternative components of the PI3K pathway promote resistance to BYL719 and identify PDK1 as the most effective co-target with PI3Kα rescuing sensitivity of resistant cells to BYL719. To explore whether PI3K pathway-independent mechanisms further contribute to BYL719 resistance, we performed phosphoproteomics and found that selection of high levels of the cell cycle regulator p21 unexpectedly promoted drug resistance in T47D cells. Functionally, high p21 levels favored repair of BYL719-induced DNA damage and bypass of the associated cellular senescence. Importantly, targeted inhibition of the check-point inhibitor CHK1 with MK-8776 effectively caused death of p21-high T47D cells, thus establishing a new vulnerability of BYL719-resistant breast cancer cells. Together, our integrated studies uncover hidden molecular mediators causing resistance to PI3Kα inhibition and provide a framework to prioritize combination therapies for PI3K-mutant breast cancer.

5.
Methods Mol Biol ; 2634: 33-58, 2023.
Article in English | MEDLINE | ID: mdl-37074573

ABSTRACT

Biochemical networks are dynamic, nonlinear, and high-dimensional systems. Realistic kinetic models of biochemical networks often comprise a multitude of kinetic parameters and state variables. Depending on the specific parameter values, a network can display one of a variety of possible dynamic behaviors such as monostable fixed point, damped oscillation, sustained oscillation, and/or bistability. Understanding how a network behaves under a particular parametric condition, and how its behavior changes as the model parameters are varied within the multidimensional parameter space are imperative for gaining a holistic understanding of the network dynamics. Such knowledge helps elucidate the parameter-to-dynamics mapping, uncover how cells make decisions under various pathophysiological contexts, and inform the design of biological circuits with desired behavior, where the latter is critical to the field of synthetic biology. In this chapter, we will present a practical guide to the multidimensional exploration, analysis, and visualization of network dynamics using pyDYVIPAC, which is a tool ideally suited to these purposes implemented in Python. The utility of pyDYVIPAC will be demonstrated using specific examples of biochemical networks with differing structures and dynamic properties via the interactive Jupyter Notebook environment.


Subject(s)
Models, Biological , Kinetics
6.
Methods Mol Biol ; 2634: 167-189, 2023.
Article in English | MEDLINE | ID: mdl-37074579

ABSTRACT

ODE modelling requires accurate knowledge of parameter and state variable values to deliver accurate and robust predictions. Parameters and state variables, however, are rarely static and immutable entities, especially in a biological context. This observation undermines the predictions made by ODE models that rely on specific parameter and state variable values and limits the contexts in which their predictions remain accurate and useful. Meta-dynamic network (MDN) modelling is a technique that can be synergistically integrated into an ODE modelling pipeline to assist in overcoming these limitations. The core mechanic of MDN modelling is the generation of a large number of model instances, each with a unique set of parameters and/or state variable values, followed by the simulation of each to determine how parameter and state variable variation affects protein dynamics. This process reveals the range of possible protein dynamics for a given network topology. Since MDN modelling is integrated with traditional ODE modelling, it can also be used to investigate the underlying causal mechanics. This technique is particularly suited to the investigation of network behaviors in systems that are highly heterogenous or systems wherein the network properties can change over time. MDN is a collection of principles rather than a strict protocol, so in this chapter, we have introduced the core principles using an example, the Hippo-ERK crosstalk signalling network.


Subject(s)
Hippo Signaling Pathway , Signal Transduction , Computer Simulation , Models, Biological
7.
Methods Mol Biol ; 2634: 357-381, 2023.
Article in English | MEDLINE | ID: mdl-37074588

ABSTRACT

The widespread development of resistance to cancer monotherapies has prompted the need to identify combinatorial treatment approaches that circumvent drug resistance and achieve more durable clinical benefit. However, given the vast space of possible combinations of existing drugs, the inaccessibility of drug screens to candidate targets with no available drugs, and the significant heterogeneity of cancers, exhaustive experimental testing of combination treatments remains highly impractical. There is thus an urgent need to develop computational approaches that complement experimental efforts and aid the identification and prioritization of effective drug combinations. Here, we provide a practical guide to SynDISCO, a computational framework that leverages mechanistic ODE modeling to predict and prioritize synergistic combination treatments directed at signaling networks. We demonstrate the key steps of SynDISCO and its application to the EGFR-MET signaling network in triple negative breast cancer as an illustrative example. SynDISCO is, however, a network- and cancer-independent framework, and given a suitable ODE model of the network of interest, it could be leveraged to discover cancer-specific combination treatments.


Subject(s)
Neoplasms , Signal Transduction , Humans , Drug Synergism , Drug Combinations , Neoplasms/drug therapy , Computational Biology
8.
Front Mol Biosci ; 10: 1094321, 2023.
Article in English | MEDLINE | ID: mdl-36743211

ABSTRACT

Precision medicine has emerged as an important paradigm in oncology, driven by the significant heterogeneity of individual patients' tumour. A key prerequisite for effective implementation of precision oncology is the development of companion biomarkers that can predict response to anti-cancer therapies and guide patient selection for clinical trials and/or treatment. However, reliable predictive biomarkers are currently lacking for many anti-cancer therapies, hampering their clinical application. Here, we developed a novel machine learning-based framework to derive predictive multi-gene biomarker panels and associated expression signatures that accurately predict cancer drug sensitivity. We demonstrated the power of the approach by applying it to identify response biomarker panels for an Hsp90-based therapy in prostate cancer, using proteomic data profiled from prostate cancer patient-derived explants. Our approach employs a rational feature section strategy to maximise model performance, and innovatively utilizes Boolean algebra methods to derive specific expression signatures of the marker proteins. Given suitable data for model training, the approach is also applicable to other cancer drug agents in different tumour settings.

9.
Oncogene ; 42(11): 833-847, 2023 03.
Article in English | MEDLINE | ID: mdl-36693952

ABSTRACT

We have determined that expression of the pseudokinase NRBP1 positively associates with poor prognosis in triple negative breast cancer (TNBC) and is required for efficient migration, invasion and proliferation of TNBC cells in culture as well as growth of TNBC orthotopic xenografts and experimental metastasis. Application of BioID/MS profiling identified P-Rex1, a known guanine nucleotide exchange factor for Rac1, as a NRBP1 binding partner. Importantly, NRBP1 overexpression enhanced levels of GTP-bound Rac1 and Cdc42 in a P-Rex1-dependent manner, while NRBP1 knockdown reduced their activation. In addition, NRBP1 associated with P-Rex1, Rac1 and Cdc42, suggesting a scaffolding function for this pseudokinase. NRBP1-mediated promotion of cell migration and invasion was P-Rex1-dependent, while constitutively-active Rac1 rescued the effect of NRBP1 knockdown on cell proliferation and invasion. Generation of reactive oxygen species via a NRBP1/P-Rex1 pathway was implicated in these oncogenic roles of NRBP1. Overall, these findings define a new function for NRBP1 and a novel oncogenic signalling pathway in TNBC that may be amenable to therapeutic intervention.


Subject(s)
Triple Negative Breast Neoplasms , Humans , rac1 GTP-Binding Protein/metabolism , Signal Transduction , Guanine Nucleotide Exchange Factors/metabolism , Cell Movement , Receptors, Cytoplasmic and Nuclear/metabolism , Vesicular Transport Proteins/metabolism
10.
J Geophys Res Space Phys ; 127(4): e2021JA030124, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35866074

ABSTRACT

Magnetosheath jets constitute a significant coupling effect between the solar wind (SW) and the magnetosphere of the Earth. In order to investigate the effects and forecasting of these jets, we present the first-ever statistical study of the jet production during large-scale SW structures like coronal mass ejections (CMEs), stream interaction regions (SIRs) and high speed streams (HSSs). Magnetosheath data from Time History of Events and Macroscale Interactions during Substorms (THEMIS) spacecraft between January 2008 and December 2020 serve as measurement source for jet detection. Two different jet definitions were used to rule out statistical biases induced by our jet detection method. For the CME and SIR + HSS lists, we used lists provided by literature and expanded on incomplete lists using OMNI data to cover the time range of May 1996 to December 2020. We find that the number and total time of observed jets decrease when CME-sheaths hit the Earth. The number of jets is lower throughout the passing of the CME-magnetic ejecta (ME) and recovers quickly afterward. On the other hand, the number of jets increases during SIR and HSS phases. We discuss a few possibilities to explain these statistical results.

11.
Am J Epidemiol ; 191(7): 1153-1173, 2022 06 27.
Article in English | MEDLINE | ID: mdl-35279711

ABSTRACT

The Collaborative Cohort of Cohorts for COVID-19 Research (C4R) is a national prospective study of adults comprising 14 established US prospective cohort studies. Starting as early as 1971, investigators in the C4R cohort studies have collected data on clinical and subclinical diseases and their risk factors, including behavior, cognition, biomarkers, and social determinants of health. C4R links this pre-coronavirus disease 2019 (COVID-19) phenotyping to information on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and acute and postacute COVID-related illness. C4R is largely population-based, has an age range of 18-108 years, and reflects the racial, ethnic, socioeconomic, and geographic diversity of the United States. C4R ascertains SARS-CoV-2 infection and COVID-19 illness using standardized questionnaires, ascertainment of COVID-related hospitalizations and deaths, and a SARS-CoV-2 serosurvey conducted via dried blood spots. Master protocols leverage existing robust retention rates for telephone and in-person examinations and high-quality event surveillance. Extensive prepandemic data minimize referral, survival, and recall bias. Data are harmonized with research-quality phenotyping unmatched by clinical and survey-based studies; these data will be pooled and shared widely to expedite collaboration and scientific findings. This resource will allow evaluation of risk and resilience factors for COVID-19 severity and outcomes, including postacute sequelae, and assessment of the social and behavioral impact of the pandemic on long-term health trajectories.


Subject(s)
COVID-19 , Adolescent , Adult , Aged , Aged, 80 and over , COVID-19/epidemiology , Cohort Studies , Humans , Middle Aged , Pandemics , Prospective Studies , SARS-CoV-2 , United States/epidemiology , Young Adult
12.
Methods Mol Biol ; 2385: 91-115, 2022.
Article in English | MEDLINE | ID: mdl-34888717

ABSTRACT

Ordinary differential equation models are used to represent intracellular signaling pathways in silico, aiding and guiding biological experiments to elucidate intracellular regulation. To construct such quantitative and predictive models of intracellular interactions, unknown parameters need to be estimated. Most of biological data are expressed in relative or arbitrary units, raising the question of how to compare model simulations with data. It has recently been shown that for models with large number of unknown parameters, fitting algorithms using a data-driven normalization of the simulations (DNS) performs best in terms of the convergence time and parameter identifiability. DNS approach compares model simulations and corresponding data both normalized by the same normalization procedure, without requiring additional parameters to be estimated, as necessary for widely used scaling factor-based methods. However, currently there is no parameter estimation software that directly supports DNS. In this chapter, we show how to apply DNS to dynamic models of systems and synthetic biology using PEPSSBI (Parameter Estimation Pipeline for Systems and Synthetic Biology). PEPSSBI is the first software that supports DNS, through algorithmically supported data normalization and objective function construction. PEPSSBI also supports model import using SBML and repeated parameter estimation runs executed in parallel either on a personal computer or a multi-CPU cluster.


Subject(s)
Models, Biological , Algorithms , Computer Simulation , Signal Transduction , Software , Systems Biology
13.
Oncogene ; 40(44): 6235-6247, 2021 11.
Article in English | MEDLINE | ID: mdl-34556814

ABSTRACT

ISG15 is an ubiquitin-like modifier that is associated with reduced survival rates in breast cancer patients. The mechanism by which ISG15 achieves this however remains elusive. We demonstrate that modification of Rab GDP-Dissociation Inhibitor Beta (GDI2) by ISG15 (ISGylation) alters endocytic recycling of the EGF receptor (EGFR) in non-interferon stimulated cells using CRISPR-knock out models for ISGylation. By regulating EGFR trafficking, ISGylation enhances EGFR recycling and sustains Akt-signalling. We further show that Akt signalling positively correlates with levels of ISG15 and its E2-ligase in basal breast cancer cohorts, confirming the link between ISGylation and Akt signalling in human tumours. Persistent and enhanced Akt activation explains the more aggressive tumour behaviour observed in human breast cancers. We show that ISGylation can act as a driver of tumour progression rather than merely being a bystander.


Subject(s)
Breast Neoplasms/metabolism , Cytokines/genetics , Cytokines/metabolism , Guanine Nucleotide Dissociation Inhibitors/metabolism , Ubiquitins/genetics , Ubiquitins/metabolism , Breast Neoplasms/genetics , CRISPR-Cas Systems , Cell Line, Tumor , Endocytosis , ErbB Receptors/metabolism , Female , Gene Knockout Techniques , Humans , Phosphorylation , Prognosis , Proteomics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Survival Analysis
14.
PLoS Comput Biol ; 17(9): e1008513, 2021 09.
Article in English | MEDLINE | ID: mdl-34529665

ABSTRACT

The PI3K/MTOR signalling network regulates a broad array of critical cellular processes, including cell growth, metabolism and autophagy. The mechanistic target of rapamycin (MTOR) kinase functions as a core catalytic subunit in two physically and functionally distinct complexes mTORC1 and mTORC2, which also share other common components including MLST8 (also known as GßL) and DEPTOR. Despite intensive research, how mTORC1 and 2 assembly and activity are coordinated, and how they are functionally linked remain to be fully characterized. This is due in part to the complex network wiring, featuring multiple feedback loops and intricate post-translational modifications. Here, we integrate predictive network modelling, in vitro experiments and -omics data analysis to elucidate the emergent dynamic behaviour of the PI3K/MTOR network. We construct new mechanistic models that encapsulate critical mechanistic details, including mTORC1/2 coordination by MLST8 (de)ubiquitination and the Akt-to-mTORC2 positive feedback loop. Model simulations validated by experimental studies revealed a previously unknown biphasic, threshold-gated dependence of mTORC1 activity on the key mTORC2 subunit SIN1, which is robust against cell-to-cell variation in protein expression. In addition, our integrative analysis demonstrates that ubiquitination of MLST8, which is reversed by OTUD7B, is regulated by IRS1/2. Our results further support the essential role of MLST8 in enabling both mTORC1 and 2's activity and suggest MLST8 as a viable therapeutic target in breast cancer. Overall, our study reports a new mechanistic model of PI3K/MTOR signalling incorporating MLST8-mediated mTORC1/2 formation and unveils a novel regulatory linkage between mTORC1 and mTORC2.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Phosphatidylinositol 3-Kinases/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Intracellular Signaling Peptides and Proteins , Mechanistic Target of Rapamycin Complex 2/chemistry , Reproducibility of Results , Signal Transduction , mTOR Associated Protein, LST8 Homolog/metabolism
15.
Elife ; 102021 07 13.
Article in English | MEDLINE | ID: mdl-34253290

ABSTRACT

The phosphoinositide 3-kinase (PI3K)-Akt network is tightly controlled by feedback mechanisms that regulate signal flow and ensure signal fidelity. A rapid overshoot in insulin-stimulated recruitment of Akt to the plasma membrane has previously been reported, which is indicative of negative feedback operating on acute timescales. Here, we show that Akt itself engages this negative feedback by phosphorylating insulin receptor substrate (IRS) 1 and 2 on a number of residues. Phosphorylation results in the depletion of plasma membrane-localised IRS1/2, reducing the pool available for interaction with the insulin receptor. Together these events limit plasma membrane-associated PI3K and phosphatidylinositol (3,4,5)-trisphosphate (PIP3) synthesis. We identified two Akt-dependent phosphorylation sites in IRS2 at S306 (S303 in mouse) and S577 (S573 in mouse) that are key drivers of this negative feedback. These findings establish a novel mechanism by which the kinase Akt acutely controls PIP3 abundance, through post-translational modification of the IRS scaffold.


For the body to work properly, cells must constantly 'talk' to each other using signalling molecules. Receiving a chemical signal triggers a series of molecular events in a cell, a so-called 'signal transduction pathway' that connects a signal with a precise outcome. Disturbing cell signalling can trigger disease, and strict control mechanisms are therefore in place to ensure that communication does not break down or become erratic. For instance, just as a thermostat turns off the heater once the right temperature is reached, negative feedback mechanisms in cells switch off signal transduction pathways when the desired outcome has been achieved. The hormone insulin is a signal for growth that increases in the body following a meal to promote the storage of excess blood glucose (sugar) in muscle and fat cells. The hormone binds to insulin receptors at the cell surface and switches on a signal transduction pathway that makes the cell take up glucose from the bloodstream. If the signal is not engaged diseases such as diabetes develop. Conversely, if the signal cannot be adequately switched of cancer can develop. Determining exactly how insulin works would help to understand these diseases better and to develop new treatments. Kearney et al. therefore set out to examine the biochemical 'fail-safes' that control insulin signalling. Experiments using computer simulations of the insulin signalling pathway revealed a potential new mechanism for negative feedback, which centred on a molecule known as Akt. The models predicted that if the negative feedback were removed, then Akt would become hyperactive and accumulate at the cell's surface after stimulation with insulin. Further manipulation of the 'virtual' insulin signalling pathway and studies of live cells in culture confirmed that this was indeed the case. The cell biology experiments also showed how Akt, once at the cell surface, was able to engage the negative feedback and shut down further insulin signalling. Akt did this by inactivating a protein required to pass the signal from the insulin receptor to the rest of the cell. Overall, this work helps to understand cell communication by revealing a previously unknown, and critical component of the insulin signalling pathway.


Subject(s)
Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/metabolism , Animals , Antigens, CD , Cell Membrane/metabolism , Computational Biology , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Receptor Substrate Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mice , Phosphorylation , Signal Transduction/physiology
16.
Int J Mol Sci ; 22(13)2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34203293

ABSTRACT

The PI3K/mTOR signalling pathway plays a central role in the governing of cell growth, survival and metabolism. As such, it must integrate and decode information from both external and internal sources to guide efficient decision-making by the cell. To facilitate this, the pathway has evolved an intricate web of complex regulatory mechanisms and elaborate crosstalk with neighbouring signalling pathways, making it a highly non-linear system. Here, we describe the mechanistic biological details that underpin these regulatory mechanisms, covering a multitude of negative and positive feedback loops, feed-forward loops, competing protein interactions, and crosstalk with major signalling pathways. Further, we highlight the non-linear and dynamic network behaviours that arise from these regulations, uncovered through computational and experimental studies. Given the pivotal role of the PI3K/mTOR network in cellular homeostasis and its frequent dysregulation in pathologies including cancer and diabetes, a coherent and systems-level understanding of the complex regulation and consequential dynamic signalling behaviours within this network is imperative for advancing biology and development of new therapeutic approaches.


Subject(s)
Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Animals , Homeostasis , Humans , Neoplasms/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
17.
iScience ; 24(2): 102118, 2021 Feb 19.
Article in English | MEDLINE | ID: mdl-33659881

ABSTRACT

Insulin's activation of PI3K/Akt signaling, stimulates glucose uptake by enhancing delivery of GLUT4 to the cell surface. Here we examined the origins of intercellular heterogeneity in insulin signaling. Akt activation alone accounted for ~25% of the variance in GLUT4, indicating that additional sources of variance exist. The Akt and GLUT4 responses were highly reproducible within the same cell, suggesting the variance is between cells (extrinsic) and not within cells (intrinsic). Generalized mechanistic models (supported by experimental observations) demonstrated that the correlation between the steady-state levels of two measured signaling processes decreases with increasing distance from each other and that intercellular variation in protein expression (as an example of extrinsic variance) is sufficient to account for the variance in and between Akt and GLUT4. Thus, the response of a population to insulin signaling is underpinned by considerable single-cell heterogeneity that is largely driven by variance in gene/protein expression between cells.

18.
J Geophys Res Space Phys ; 126(11): e2021JA029770, 2021 Nov.
Article in English | MEDLINE | ID: mdl-35864948

ABSTRACT

One of the grand challenges in heliophysics is the characterization of coronal mass ejection (CME) magnetic structure and evolution from eruption at the Sun through heliospheric propagation. At present, the main difficulties are related to the lack of direct measurements of the coronal magnetic fields and the lack of 3D in-situ measurements of the CME body in interplanetary space. Nevertheless, the evolution of a CME magnetic structure can be followed using a combination of multi-point remote-sensing observations and multi-spacecraft in-situ measurements as well as modeling. Accordingly, we present in this work the analysis of two CMEs that erupted from the Sun on April 28, 2012. We follow their eruption and early evolution using remote-sensing data, finding indications of CME-CME interaction, and then analyze their interplanetary counterpart(s) using in-situ measurements at Venus, Earth, and Saturn. We observe a seemingly single flux rope at all locations, but find possible signatures of interaction at Earth, where high-cadence plasma data are available. Reconstructions of the in-situ flux ropes provide almost identical results at Venus and Earth but show greater discrepancies at Saturn, suggesting that the CME was highly distorted and/or that further interaction with nearby solar wind structures took place before 10 AU. This work highlights the difficulties in connecting structures from the Sun to the outer heliosphere and demonstrates the importance of multi-spacecraft studies to achieve a deeper understanding of the magnetic configuration of CMEs.

19.
Mol Cell ; 80(2): 279-295.e8, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33065020

ABSTRACT

The PTEN tumor suppressor controls cell death and survival by regulating functions of various molecular targets. While the role of PTEN lipid-phosphatase activity on PtdIns(3,4,5)P3 and inhibition of PI3K pathway is well characterized, the biological relevance of PTEN protein-phosphatase activity remains undefined. Here, using knockin (KI) mice harboring cancer-associated and functionally relevant missense mutations, we show that although loss of PTEN lipid-phosphatase function cooperates with oncogenic PI3K to promote rapid mammary tumorigenesis, the additional loss of PTEN protein-phosphatase activity triggered an extensive cell death response evident in early and advanced mammary tumors. Omics and drug-targeting studies revealed that PI3Ks act to reduce glucocorticoid receptor (GR) levels, which are rescued by loss of PTEN protein-phosphatase activity to restrain cell survival. Thus, we find that the dual regulation of GR by PI3K and PTEN functions as a rheostat that can be exploited for the treatment of PTEN loss-driven cancers.


Subject(s)
Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , PTEN Phosphohydrolase/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Carcinogenesis , Cell Death , Cell Line, Tumor , Cell Proliferation , Dexamethasone/pharmacology , Female , Humans , Isoenzymes/metabolism , Mice , Models, Biological , Mutation/genetics , Organoids/pathology , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Protein Stability , Proteome/metabolism , Proto-Oncogene Proteins c-akt/metabolism
20.
Mol Cell Proteomics ; 19(11): 1777-1789, 2020 11.
Article in English | MEDLINE | ID: mdl-32759169

ABSTRACT

Amino acid hydroxylation is a common post-translational modification, which generally regulates protein interactions or adds a functional group that can be further modified. Such hydroxylation is currently considered irreversible, necessitating the degradation and re-synthesis of the entire protein to reset the modification. Here we present evidence that the cellular machinery can reverse FIH-mediated asparagine hydroxylation on intact proteins. These data suggest that asparagine hydroxylation is a flexible and dynamic post-translational modification akin to modifications involved in regulating signaling networks, such as phosphorylation, methylation and ubiquitylation.


Subject(s)
Asparagine/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mixed Function Oxygenases/metabolism , Protein Processing, Post-Translational , Repressor Proteins/metabolism , TRPV Cation Channels/metabolism , Tankyrases/metabolism , Amino Acid Sequence , Cell Line, Tumor , Humans , Hydroxylation , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Kinetics , Mass Spectrometry , Methylation , Mixed Function Oxygenases/genetics , Phosphorylation , Protein Binding , Repressor Proteins/genetics , Signal Transduction , TRPV Cation Channels/genetics , Tankyrases/genetics , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...