Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
J Eval Clin Pract ; 30(2): 184-198, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37721181

ABSTRACT

RATIONALE: Delivering optimal patient health care requires interdisciplinary clinician communication. A single communication tool across multiple pre-hospital and hospital settings, and between hospital departments is a novel solution to current systems. Fit-for-purpose, secure smartphone applications allow clinical information to be shared quickly between health providers. Little is known as to what underpins their successful implementation in an emergency care context. AIMS: To identify (a) whether implementing a single, digital health communication application across multiple health care organisations and hospital departments is feasible; (b) the barriers and facilitators to implementation; and (c) which factors are associated with clinicians' intentions to use the technology. METHODS: We used a multimethod design, evaluating the implementation of a secure, digital communication application (Pulsara™). The technology was trialled in two Australian regional hospitals and 25 Ambulance Victoria branches (AV). Post-training, clinicians involved in treating patients with suspected stroke or cardiac events were administered surveys measuring perceived organisational readiness (Organisational Readiness for Implementing Change), clinicians' intentions (Unified Theory of Acceptance and Use of Technology) and internal motivations (Self-Determination Theory) to use Pulsara™, and the perceived benefits and barriers of use. Quantitative data were descriptively summarised with multivariable associations between factors and intentions to use Pulsara™ examined with linear regression. Qualitative data responses were subjected to directed content analysis (two coders). RESULTS: Participants were paramedics (n = 82, median 44 years) or hospital-based clinicians (n = 90, median 37 years), with organisations perceived to be similarly ready. Regression results (F(11, 136) = 21.28, p = <0.001, Adj R2 = 0.60) indicated Habit, Effort Expectancy, Perceived Organisational Readiness, Performance Expectancy and Organisation membership (AV) as predictors of intending to use Pulsara™. Themes relating to benefits (95% coder agreement) included improved communication, procedural efficiencies and faster patient care. Barriers (92% coder agreement) included network accessibility and remembering passwords. PulsaraTM was initiated 562 times. CONCLUSION: Implementing multiorganisational, digital health communication applications is feasible, and facilitated when organisations are change-ready for an easy-to-use, effective solution. Developing habitual use is key, supported through implementation strategies (e.g., hands-on training). Benefits should be emphasised (e.g., during education sessions), including streamlining communication and patient flow, and barriers addressed (e.g., identify champions and local technical support) at project commencement.


Subject(s)
Emergency Medical Services , Interdisciplinary Communication , Humans , Digital Health , Australia , Delivery of Health Care
2.
J Pathol ; 258(4): 382-394, 2022 12.
Article in English | MEDLINE | ID: mdl-36073856

ABSTRACT

PTEN is one of the most commonly inactivated tumour suppressor genes in sporadic cancer. Germline heterozygous PTEN gene alterations also underlie PTEN hamartoma tumour syndrome (PHTS), a rare human cancer-predisposition condition. A key feature of systemic PTEN deregulation is the inability to adequately dampen PI3-kinase (PI3K)/mTORC1 signalling. PI3K/mTORC1 pathway inhibitors such as rapamycin are therefore expected to neutralise the impact of PTEN loss, rendering this a more druggable context compared with those of other tumour suppressor pathways such as loss of TP53. However, this has not been explored in cancer prevention in a model of germline cancer predisposition, such as PHTS. Clinical trials of short-term treatment with rapamycin have recently been initiated for PHTS, focusing on cognition and colon polyposis. Here, we administered a low dose of rapamycin from the age of 6 weeks onwards to mice with heterozygous germline Pten loss, a mouse model that recapitulates most characteristics of human PHTS. Rapamycin was well tolerated and led to a highly significant improvement of survival in both male and female mice. This was accompanied by a delay in, but not full blockade of, the development of a range of proliferative lesions, including gastro-intestinal and thyroid tumours and endometrial hyperplasia, with no impact on mammary and prostate tumours, and no effect on brain overgrowth. Our data indicate that rapamycin may have cancer prevention potential in human PHTS. This might also be the case for sporadic cancers in which genetic PI3K pathway activation is an early event in tumour development, such as endometrial cancer and some breast cancers. To the best of our knowledge, this is the first report of a long-term treatment of a germline cancer predisposition model with a PI3K/mTOR pathway inhibitor. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Hamartoma Syndrome, Multiple , Thyroid Neoplasms , Mice , Animals , Male , Female , Humans , Infant , Sirolimus/pharmacology , Sirolimus/therapeutic use , Phosphatidylinositol 3-Kinases/genetics , Longevity , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Hamartoma Syndrome, Multiple/drug therapy , Hamartoma Syndrome, Multiple/genetics , Hamartoma Syndrome, Multiple/pathology , Phosphatidylinositol 3-Kinase/genetics , Phosphoinositide-3 Kinase Inhibitors , Mechanistic Target of Rapamycin Complex 1/genetics , Germ Cells/metabolism , Germ-Line Mutation
3.
BMJ Open ; 12(7): e052332, 2022 07 18.
Article in English | MEDLINE | ID: mdl-35851025

ABSTRACT

OBJECTIVES: To determine if a digital communication app improves care timelines for patients with suspected acute stroke/ST-elevation myocardial infarction (STEMI). DESIGN: Real-world feasibility study, quasi-experimental design. SETTING: Prehospital (25 Ambulance Victoria branches) and within-hospital (2 hospitals) in regional Victoria, Australia. PARTICIPANTS: Paramedics or emergency department (ED) clinicians identified patients with suspected acute stroke (onset <4.5 hours; n=604) or STEMI (n=247). INTERVENTION: The Pulsara communication app provides secure, two-way, real-time communication. Assessment and treatment times were recorded for 12 months (May 2017-April 2018), with timelines compared between 'Pulsara initiated' (Pulsara) and 'not initiated' (no Pulsara). PRIMARY OUTCOME MEASURE: Door-to-treatment (needle for stroke, balloon for STEMI) Secondary outcome measures: ambulance and hospital processes. RESULTS: Stroke (no Pulsara n=215, Pulsara n=389) and STEMI (no Pulsara n=76, Pulsara n=171) groups were of similar age and sex (stroke: 76 vs 75 years; both groups 50% male; STEMI: 66 vs 63 years; 68% and 72% male). When Pulsara was used, patients were off ambulance stretcher faster for stroke (11(7, 17) vs 19(11, 29); p=0.0001) and STEMI (14(7, 23) vs 19(10, 32); p=0.0014). ED door-to-first medical review was faster (6(2, 14) vs 23(8, 67); p=0.0001) for stroke but only by 1 min for STEMI (3 (0, 7) vs 4 (0, 14); p=0.25). Door-to-CT times were 44 min faster (27(18, 44) vs 71(43, 147); p=0.0001) for stroke, and percutaneous intervention door-to-balloon times improved by 17 min, but non-significant (56 (34, 88) vs 73 (49, 110); p=0.41) for STEMI. There were improvements in the proportions of patients treated within 60 min for stroke (12%-26%, p=0.15) and 90 min for STEMI (50%-78%, p=0.20). CONCLUSIONS: In this Australian-first study, uptake of the digital communication app was strong, patient-centred care timelines improved, although door-to-treatment times remained similar.


Subject(s)
Emergency Medical Services , Mobile Applications , Myocardial Infarction , ST Elevation Myocardial Infarction , Stroke , Ambulances , Arrhythmias, Cardiac , Communication , Electrocardiography , Feasibility Studies , Female , Humans , Male , Myocardial Infarction/therapy , ST Elevation Myocardial Infarction/therapy , Stroke/therapy , Time Factors , Treatment Outcome , Victoria
4.
Nat Commun ; 12(1): 117, 2021 01 05.
Article in English | MEDLINE | ID: mdl-33402692

ABSTRACT

Nasopharyngeal cancer (NPC), endemic in Southeast Asia, lacks effective diagnostic and therapeutic strategies. Even in high-income countries the 5-year survival rate for stage IV NPC is less than 40%. Here we report high somatostatin receptor 2 (SSTR2) expression in multiple clinical cohorts comprising 402 primary, locally recurrent and metastatic NPCs. We show that SSTR2 expression is induced by the Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) via the NF-κB pathway. Using cell-based and preclinical rodent models, we demonstrate the therapeutic potential of SSTR2 targeting using a cytotoxic drug conjugate, PEN-221, which is found to be superior to FDA-approved SSTR2-binding cytostatic agents. Furthermore, we reveal significant correlation of SSTR expression with increased rates of survival and report in vivo uptake of the SSTR2-binding 68Ga-DOTA-peptide radioconjugate in PET-CT scanning in a clinical trial of NPC patients (NCT03670342). These findings reveal a key role in EBV-associated NPC for SSTR2 in infection, imaging, targeted therapy and survival.


Subject(s)
Epstein-Barr Virus Infections , Gene Expression Regulation, Neoplastic , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Neoplasm Recurrence, Local , Receptors, Somatostatin , Viral Matrix Proteins , Animals , Female , Humans , Male , Mice , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/mortality , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/pathogenicity , Host-Pathogen Interactions/genetics , Lymphatic Metastasis , Mice, Nude , Molecular Targeted Therapy , Nasopharyngeal Carcinoma/drug therapy , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/mortality , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/mortality , Nasopharyngeal Neoplasms/virology , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/virology , NF-kappa B/genetics , NF-kappa B/metabolism , Octreotide/pharmacology , Positron Emission Tomography Computed Tomography , Receptors, Somatostatin/antagonists & inhibitors , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism , Signal Transduction , Survival Analysis , Viral Matrix Proteins/antagonists & inhibitors , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism , Xenograft Model Antitumor Assays
5.
Proc Natl Acad Sci U S A ; 116(17): 8380-8389, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30948643

ABSTRACT

The PIK3CA gene, which encodes the p110α catalytic subunit of PI3 kinase (PI3K), is mutationally activated in cancer and in overgrowth disorders known as PIK3CA-related overgrowth spectrum (PROS). To determine the consequences of genetic PIK3CA activation in a developmental context of relevance to both PROS and cancer, we engineered isogenic human induced pluripotent stem cells (iPSCs) with heterozygous or homozygous knockin of PIK3CAH1047R While heterozygous iPSCs remained largely similar to wild-type cells, homozygosity for PIK3CAH1047R caused widespread, cancer-like transcriptional remodeling, partial loss of epithelial morphology, up-regulation of stemness markers, and impaired differentiation to all three germ layers in vitro and in vivo. Genetic analysis of PIK3CA-associated cancers revealed that 64% had multiple oncogenic PIK3CA copies (39%) or additional PI3K signaling pathway-activating "hits" (25%). This contrasts with the prevailing view that PIK3CA mutations occur heterozygously in cancer. Our findings suggest that a PI3K activity threshold determines pathological consequences of oncogenic PIK3CA activation and provide insight into the specific role of this pathway in human pluripotent stem cells.


Subject(s)
Class I Phosphatidylinositol 3-Kinases , Induced Pluripotent Stem Cells , Neoplasms , Adolescent , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Class I Phosphatidylinositol 3-Kinases/physiology , Female , Gene Editing , Gene Expression Regulation, Neoplastic/genetics , Gene Knock-In Techniques , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/physiology , Male , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction/genetics
7.
Nat Commun ; 8(1): 1804, 2017 11 27.
Article in English | MEDLINE | ID: mdl-29180704

ABSTRACT

Vps34 PI3K is thought to be the main producer of phosphatidylinositol-3-monophosphate, a lipid that controls intracellular vesicular trafficking. The organismal impact of systemic inhibition of Vps34 kinase activity is not completely understood. Here we show that heterozygous Vps34 kinase-dead mice are healthy and display a robustly enhanced insulin sensitivity and glucose tolerance, phenotypes mimicked by a selective Vps34 inhibitor in wild-type mice. The underlying mechanism of insulin sensitization is multifactorial and not through the canonical insulin/Akt pathway. Vps34 inhibition alters cellular energy metabolism, activating the AMPK pathway in liver and muscle. In liver, Vps34 inactivation mildly dampens autophagy, limiting substrate availability for mitochondrial respiration and reducing gluconeogenesis. In muscle, Vps34 inactivation triggers a metabolic switch from oxidative phosphorylation towards glycolysis and enhanced glucose uptake. Our study identifies Vps34 as a new drug target for insulin resistance in Type-2 diabetes, in which the unmet therapeutic need remains substantial.


Subject(s)
Insulin Resistance , Mitochondria/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/physiology , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy/physiology , Cell Line, Tumor , Class III Phosphatidylinositol 3-Kinases , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Gene Knock-In Techniques , Glucose/analysis , Glucose/metabolism , Glucose Tolerance Test , Glycolysis/physiology , Hepatocytes , Heterozygote , Humans , Insulin/metabolism , Liver/cytology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myoblasts , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Primary Cell Culture
8.
Nat Commun ; 8(1): 1773, 2017 11 24.
Article in English | MEDLINE | ID: mdl-29170395

ABSTRACT

Mutations in PIK3CA are very frequent in cancer and lead to sustained PI3K pathway activation. The impact of acute expression of mutant PIK3CA during early stages of malignancy is unknown. Using a mouse model to activate the Pik3ca H1047R hotspot mutation in the heterozygous state from its endogenous locus, we here report that mutant Pik3ca induces centrosome amplification in cultured cells (through a pathway involving AKT, ROCK and CDK2/Cyclin E-nucleophosmin) and in mouse tissues, and increased in vitro cellular tolerance to spontaneous genome doubling. We also present evidence that the majority of PIK3CA H1047R mutations in the TCGA breast cancer cohort precede genome doubling. These previously unappreciated roles of PIK3CA mutation show that PI3K signalling can contribute to the generation of irreversible genomic changes in cancer. While this can limit the impact of PI3K-targeted therapies, these findings also open the opportunity for therapeutic approaches aimed at limiting tumour heterogeneity and evolution.


Subject(s)
Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Centrosome/metabolism , Class I Phosphatidylinositol 3-Kinases/metabolism , Gene Amplification , Genome , Phosphatidylinositol 3-Kinases/metabolism , Animals , Class I Phosphatidylinositol 3-Kinases/genetics , Cohort Studies , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mutation , Oncogenes , Phosphatidylinositol 3-Kinases/genetics
9.
Diabetologia ; 59(7): 1503-1512, 2016 07.
Article in English | MEDLINE | ID: mdl-27138914

ABSTRACT

AIMS/HYPOTHESIS: While the class I phosphoinositide 3-kinases (PI3Ks) are well-documented positive regulators of metabolism, the involvement of class II PI3K isoforms (PI3K-C2α, -C2ß and -C2γ) in metabolic regulation is just emerging. Organismal inactivation of PI3K-C2ß increases insulin signalling and sensitivity, whereas PI3K-C2γ inactivation has a negative metabolic impact. In contrast, the role of PI3K-C2α in organismal metabolism remains unexplored. In this study, we investigated whether kinase inactivation of PI3K-C2α affects glucose metabolism in mice. METHODS: We have generated and characterised a mouse line with a constitutive inactivating knock-in (KI) mutation in the kinase domain of the gene encoding PI3K-C2α (Pik3c2a). RESULTS: While homozygosity for kinase-dead PI3K-C2α was embryonic lethal, heterozygous PI3K-C2α KI mice were viable and fertile, with no significant histopathological findings. However, male heterozygous mice showed early onset leptin resistance, with a defect in leptin signalling in the hypothalamus, correlating with a mild, age-dependent obesity, insulin resistance and glucose intolerance. Insulin signalling was unaffected in insulin target tissues of PI3K-C2α KI mice, in contrast to previous reports in which downregulation of PI3K-C2α in cell lines was shown to dampen insulin signalling. Interestingly, no metabolic phenotypes were detected in female PI3K-C2α KI mice at any age. CONCLUSIONS/INTERPRETATION: Our data uncover a sex-dependent role for PI3K-C2α in the modulation of hypothalamic leptin action and systemic glucose homeostasis. ACCESS TO RESEARCH MATERIALS: All reagents are available upon request.


Subject(s)
Insulin Resistance/physiology , Leptin/metabolism , Obesity/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Adipocytes/metabolism , Animals , Blotting, Western , Cell Line , Eating/genetics , Eating/physiology , Glucose/metabolism , Homeostasis/genetics , Homeostasis/physiology , Hypothalamus/metabolism , Insulin/metabolism , Insulin Resistance/genetics , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Phosphatidylinositol 3-Kinases/genetics , Signal Transduction/genetics , Signal Transduction/physiology
11.
Sci Transl Med ; 8(332): 332ra43, 2016 Mar 30.
Article in English | MEDLINE | ID: mdl-27030595

ABSTRACT

Venous malformations (VMs) are painful and deforming vascular lesions composed of dilated vascular channels, which are present from birth. Mutations in the TEK gene, encoding the tyrosine kinase receptor TIE2, are found in about half of sporadic (nonfamilial) VMs, and the causes of the remaining cases are unknown. Sclerotherapy, widely accepted as first-line treatment, is not fully efficient, and targeted therapy for this disease remains underexplored. We have generated a mouse model that faithfully mirrors human VM through mosaic expression of Pik3ca(H1047R), a constitutively active mutant of the p110α isoform of phosphatidylinositol 3-kinase (PI3K), in the embryonic mesoderm. Endothelial expression of Pik3ca(H1047R)resulted in endothelial cell (EC) hyperproliferation, reduction in pericyte coverage of blood vessels, and decreased expression of arteriovenous specification markers. PI3K pathway inhibition with rapamycin normalized EC hyperproliferation and pericyte coverage in postnatal retinas and stimulated VM regression in vivo. In line with the mouse data, we also report the presence of activating PIK3CA mutations in human VMs, mutually exclusive with TEK mutations. Our data demonstrate a causal relationship between activating Pik3ca mutations and the genesis of VMs, provide a genetic model that faithfully mirrors the normal etiology and development of this human disease, and establish the basis for the use of PI3K-targeted therapies in VMs.


Subject(s)
Mutation/genetics , Phosphatidylinositol 3-Kinases/genetics , Vascular Malformations/enzymology , Vascular Malformations/genetics , Animals , Cell Proliferation/drug effects , Class I Phosphatidylinositol 3-Kinases , Endothelial Cells/drug effects , Endothelial Cells/pathology , Humans , Mesoderm/drug effects , Mesoderm/embryology , Mesoderm/pathology , Mice, Inbred C57BL , Mosaicism/drug effects , Pericytes/drug effects , Pericytes/pathology , Receptor, TIE-2/metabolism , Sirolimus/pharmacology
12.
Cell Rep ; 13(9): 1881-94, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26655903

ABSTRACT

In contrast to the class I phosphoinositide 3-kinases (PI3Ks), the organismal roles of the kinase activity of the class II PI3Ks are less clear. Here, we report that class II PI3K-C2ß kinase-dead mice are viable and healthy but display an unanticipated enhanced insulin sensitivity and glucose tolerance, as well as protection against high-fat-diet-induced liver steatosis. Despite having a broad tissue distribution, systemic PI3K-C2ß inhibition selectively enhances insulin signaling only in metabolic tissues. In a primary hepatocyte model, basal PI3P lipid levels are reduced by 60% upon PI3K-C2ß inhibition. This results in an expansion of the very early APPL1-positive endosomal compartment and altered insulin receptor trafficking, correlating with an amplification of insulin-induced, class I PI3K-dependent Akt signaling, without impacting MAPK activity. These data reveal PI3K-C2ß as a critical regulator of endosomal trafficking, specifically in insulin signaling, and identify PI3K-C2ß as a potential drug target for insulin sensitization.


Subject(s)
Class II Phosphatidylinositol 3-Kinases/metabolism , Insulin/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagy , Blood Glucose/analysis , Cells, Cultured , Class II Phosphatidylinositol 3-Kinases/genetics , Diet, High-Fat , Endosomes/metabolism , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Knock-In Techniques , Hepatocytes/cytology , Hepatocytes/metabolism , Insulin/blood , Liver/pathology , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
13.
PLoS Genet ; 11(7): e1005304, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26132308

ABSTRACT

The organismal roles of the ubiquitously expressed class I PI3K isoform p110ß remain largely unknown. Using a new kinase-dead knockin mouse model that mimics constitutive pharmacological inactivation of p110ß, we document that full inactivation of p110ß leads to embryonic lethality in a substantial fraction of mice. Interestingly, the homozygous p110ß kinase-dead mice that survive into adulthood (maximum ~26% on a mixed genetic background) have no apparent phenotypes, other than subfertility in females and complete infertility in males. Systemic inhibition of p110ß results in a highly specific blockade in the maturation of spermatogonia to spermatocytes. p110ß was previously suggested to signal downstream of the c-kit tyrosine kinase receptor in germ cells to regulate their proliferation and survival. We now report that p110ß also plays a germ cell-extrinsic role in the Sertoli cells (SCs) that support the developing sperm, with p110ß inactivation dampening expression of the SC-specific Androgen Receptor (AR) target gene Rhox5, a homeobox gene critical for spermatogenesis. All extragonadal androgen-dependent functions remain unaffected by global p110ß inactivation. In line with a crucial role for p110ß in SCs, selective inactivation of p110ß in these cells results in male infertility. Our study is the first documentation of the involvement of a signalling enzyme, PI3K, in the regulation of AR activity during spermatogenesis. This developmental pathway may become active in prostate cancer where p110ß and AR have previously been reported to functionally interact.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , Fertility/physiology , Infertility, Male/genetics , Receptors, Androgen/metabolism , Sertoli Cells/metabolism , Animals , Blastocyst/cytology , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Class I Phosphatidylinositol 3-Kinases/genetics , Female , Homeodomain Proteins/genetics , Infertility, Female/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Morula/cytology , Receptors, Androgen/genetics , Signal Transduction/genetics , Spermatogenesis/genetics , Transcription Factors/genetics , Transcription, Genetic/genetics
14.
Nature ; 510(7505): 407-411, 2014 06 19.
Article in English | MEDLINE | ID: mdl-24919154

ABSTRACT

Inhibitors against the p110δ isoform of phosphoinositide-3-OH kinase (PI(3)K) have shown remarkable therapeutic efficacy in some human leukaemias. As p110δ is primarily expressed in leukocytes, drugs against p110δ have not been considered for the treatment of solid tumours. Here we report that p110δ inactivation in mice protects against a broad range of cancers, including non-haematological solid tumours. We demonstrate that p110δ inactivation in regulatory T cells unleashes CD8(+) cytotoxic T cells and induces tumour regression. Thus, p110δ inhibitors can break tumour-induced immune tolerance and should be considered for wider use in oncology.


Subject(s)
Enzyme Inhibitors/pharmacology , Immune Tolerance/drug effects , Neoplasms/enzymology , Neoplasms/immunology , Phosphatidylinositol 3-Kinases/metabolism , T-Lymphocytes, Regulatory/drug effects , Animals , Antineoplastic Agents/pharmacology , Enzyme Activation/drug effects , Immune Tolerance/immunology , Mice , T-Lymphocytes, Regulatory/enzymology , T-Lymphocytes, Regulatory/immunology
15.
J Exp Med ; 210(10): 1937-45, 2013 Sep 23.
Article in English | MEDLINE | ID: mdl-24043760

ABSTRACT

Understanding the direct, tumor cell-intrinsic effects of PI 3-kinase (PI3K) has been a key focus of research to date. Here, we report that cancer cell-extrinsic PI3K activity, mediated by the p110α isoform of PI3K, contributes in an unexpected way to tumor angiogenesis. In syngeneic mouse models, inactivation of stromal p110α led to increased vascular density, reduced vessel size, and altered pericyte coverage. This increased vascularity lacked functionality, correlating with enhanced tumor hypoxia and necrosis, and reduced tumor growth. The role of p110α in tumor angiogenesis is multifactorial, and includes regulation of proliferation and DLL4 expression in endothelial cells. p110α in the tumor stroma is thus a regulator of vessel formation, with p110α inactivation giving rise to nonfunctional angiogenesis, which can stunt tumor growth. This type of vascular aberration differs from vascular endothelial growth factor-centered antiangiogenesis therapies, which mainly lead to vascular pruning. Inhibition of p110α may thus offer a new antiangiogenic therapeutic opportunity in cancer.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Class I Phosphatidylinositol 3-Kinases , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Melanoma, Experimental , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Neoplasms/genetics , Neovascularization, Pathologic/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Stromal Cells/drug effects , Stromal Cells/metabolism
16.
EMBO Mol Med ; 5(4): 563-71, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23483710

ABSTRACT

The insulin/insulin-like growth factor-1 signalling (IIS) pathway regulates cellular and organismal metabolism and controls the rate of aging. Gain-of-function mutations in p110α, the principal mammalian IIS-responsive isoform of PI 3-kinase (PI3K), promote cancer. In contrast, loss-of-function mutations in p110α impair insulin signalling and cause insulin resistance, inducing a pre-diabetic state. It remains unknown if long-term p110α inactivation induces further metabolic deterioration over time, leading to overt unsustainable pathology. Surprisingly, we find that chronic p110α partial inactivation in mice protects from age-related reduction in insulin sensitivity, glucose tolerance and fat accumulation, and extends the lifespan of male mice. This beneficial effect of p110α inactivation derives in part from a suppressed down-regulation of insulin receptor substrate (IRS) protein levels induced by age-related hyperinsulinemia, and correlates with enhanced insulin-induced Akt signalling in aged p110α-deficient mice. This temporal metabolic plasticity upon p110α inactivation indicates that prolonged PI3K inhibition, as intended in human cancer treatment, might not negatively impact on organismal metabolism.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Metabolic Diseases/enzymology , Animals , Fats/metabolism , Female , Gene Silencing , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Male , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Time Factors
17.
Biochem J ; 450(3): 619-28, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23330613

ABSTRACT

PI3K (phosphoinositide 3-kinase) signalling pathways regulate a large array of cell biological functions in normal and cancer cells. In the present study we investigated the involvement of PI3K in modulating small molecule metabolism. A LC (liquid chromatography)-MS screen in colorectal cancer cell lines isogenic for oncogenic PIK3CA mutations revealed an association between PI3K activation and the levels of polyamine pathway metabolites, including 5-methylthioadenosine, putrescine and spermidine. Pharmacological inhibition confirmed that the PI3K pathway controls polyamine production. Despite inducing a decrease in PKB (protein kinase B)/Akt phosphorylation, spermidine promoted cell survival and opposed the anti-proliferative effects of PI3K inhibitors. Conversely, polyamine depletion by an ornithine decarboxylase inhibitor enhanced PKB/Akt phosphorylation, but suppressed cell survival. These results suggest that spermidine mediates cell proliferation and survival downstream of PI3K/Akt and indicate that these two biochemical pathways control each other's activities, highlighting a mechanism by which small molecule metabolism feeds back to regulate kinase signalling. Consistent with this feedback loop having a functional role in these cell models, pharmacological inhibitors of PI3K and ornithine decarboxylase potentiated each other in inhibiting tumour growth in a xenograft model. The results of the present study support the notion that the modulation of spermidine concentrations may be a previously unrecognized mechanism by which PI3K sustains chronic proliferation of cancer cells.


Subject(s)
Cell Proliferation , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Polyamines/metabolism , Animals , Chromatography, Liquid , Female , Humans , Mass Spectrometry , Metabolome , Mice , Mice, SCID , Neoplasm Transplantation , Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/physiology , Transplantation, Heterologous , Tumor Cells, Cultured
18.
Nat Immunol ; 13(11): 1045-1054, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23023391

ABSTRACT

Lipopolysaccharide activates plasma-membrane signaling and endosomal signaling by Toll-like receptor 4 (TLR4) through the TIRAP-MyD88 and TRAM-TRIF adaptor complexes, respectively, but it is unclear how the signaling switch between these cell compartments is coordinated. In dendritic cells, we found that the p110δ isoform of phosphatidylinositol-3-OH kinase (PI(3)K) induced internalization of TLR4 and dissociation of TIRAP from the plasma membrane, followed by calpain-mediated degradation of TIRAP. Accordingly, inactivation of p110δ prolonged TIRAP-mediated signaling from the plasma membrane, which augmented proinflammatory cytokine production while decreasing TRAM-dependent endosomal signaling that generated anti-inflammatory cytokines (interleukin 10 and interferon-ß). In line with that altered signaling output, p110δ-deficient mice showed enhanced endotoxin-induced death. Thus, by controlling the 'topology' of TLR4 signaling complexes, p110δ balances overall homeostasis in the TLR4 pathway.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/immunology , Dendritic Cells/immunology , Signal Transduction/immunology , Toll-Like Receptor 4/immunology , Animals , Calpain/pharmacology , Cell Compartmentation/immunology , Cell Membrane/drug effects , Cell Membrane/genetics , Cell Membrane/immunology , Cells, Cultured , Class Ia Phosphatidylinositol 3-Kinase/genetics , Dendritic Cells/cytology , Dendritic Cells/drug effects , Endosomes/drug effects , Endosomes/genetics , Endosomes/immunology , Gene Expression/drug effects , Gene Expression/immunology , Interferon-beta/biosynthesis , Interferon-beta/immunology , Interleukin-10/biosynthesis , Interleukin-10/immunology , Isoenzymes/genetics , Isoenzymes/immunology , Lipopolysaccharides/pharmacology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Receptors, Interleukin/genetics , Receptors, Interleukin/immunology , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/immunology , Shock, Septic/genetics , Shock, Septic/immunology , Signal Transduction/drug effects , Signal Transduction/genetics , Toll-Like Receptor 4/genetics
19.
Biochem J ; 442(1): 151-9, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22150431

ABSTRACT

The PI3K (phosphoinositide 3-kinase) pathway is commonly activated in cancer as a consequence of inactivation of the tumour suppressor PTEN (phosphatase and tensin homologue deleted on chromosome 10), a major negative regulator of PI3K signalling. In line with this important role of PTEN, mice that are heterozygous for a PTEN-null allele (PTEN+/− mice) spontaneously develop a variety of tumours in multiple organs. PTEN is a phosphatase with selectivity for PtdIns(3,4,5)P3, which is produced by the class I isoforms of PI3K (p110α, p110ß, p110γ and p110δ). Previous studies indicated that PTEN-deficient cancer cell lines mainly depend on p110ß, and that p110ß, but not p110α, controls mouse prostate cancer development driven by PTEN loss. In the present study, we investigated whether the ubiquitously expressed p110α can also functionally interact with PTEN in cancer. Using genetic mouse models that mimic systemic administration of p110α- or p110ß-selective inhibitors, we confirm that inactivation of p110ß, but not p110α, inhibits prostate cancer development in PTEN+/− mice, but also find that p110α inactivation protects from glomerulonephritis, pheochromocytoma and thyroid cancer induced by PTEN loss. This indicates that p110α can modulate the impact of PTEN loss in disease and tumourigenesis. In primary and immortalized mouse fibroblast cell lines, both p110α and p110ß controlled steady-state PtdIns(3,4,5)P3 levels and Akt signalling induced by heterozygous PTEN loss. In contrast, no correlation was found in primary mouse tissues between PtdIns(3,4,5)P3 levels, PI3K/PTEN genotype and cancer development. Taken together, our results from the present study show that inactivation of either p110α or p110ß can counteract the impact of PTEN inactivation. The potential implications of these findings for PI3K-targeted therapy of cancer are discussed.


Subject(s)
PTEN Phosphohydrolase/antagonists & inhibitors , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cell Transformation, Neoplastic , Class I Phosphatidylinositol 3-Kinases , Isoenzymes/metabolism , Lymphoma/etiology , Male , Mice , Phosphoinositide-3 Kinase Inhibitors , Prostatic Neoplasms/etiology , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/physiology , Thyroid Neoplasms/prevention & control
20.
Nature ; 453(7195): 662-6, 2008 May 29.
Article in English | MEDLINE | ID: mdl-18449193

ABSTRACT

Phosphoinositide 3-kinases (PI3Ks) signal downstream of multiple cell-surface receptor types. Class IA PI3K isoforms couple to tyrosine kinases and consist of a p110 catalytic subunit (p110alpha, p110beta or p110delta), constitutively bound to one of five distinct p85 regulatory subunits. PI3Ks have been implicated in angiogenesis, but little is known about potential selectivity among the PI3K isoforms and their mechanism of action in endothelial cells during angiogenesis in vivo. Here we show that only p110alpha activity is essential for vascular development. Ubiquitous or endothelial cell-specific inactivation of p110alpha led to embryonic lethality at mid-gestation because of severe defects in angiogenic sprouting and vascular remodelling. p110alpha exerts this critical endothelial cell-autonomous function by regulating endothelial cell migration through the small GTPase RhoA. p110alpha activity is particularly high in endothelial cells and preferentially induced by tyrosine kinase ligands (such as vascular endothelial growth factor (VEGF)-A). In contrast, p110beta in endothelial cells signals downstream of G-protein-coupled receptor (GPCR) ligands such as SDF-1alpha, whereas p110delta is expressed at low level and contributes only minimally to PI3K activity in endothelial cells. These results provide the first in vivo evidence for p110-isoform selectivity in endothelial PI3K signalling during angiogenesis.


Subject(s)
Cell Movement , Endothelial Cells/cytology , Endothelial Cells/enzymology , Neovascularization, Physiologic , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cells, Cultured , Class I Phosphatidylinositol 3-Kinases , Female , Humans , Mice , Phosphatidylinositol 3-Kinases/genetics , RNA Interference , Rats , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Wounds and Injuries , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...