1
|
Merz LM, Winter K, Richter S, Kallendrusch S, Horn A, Grunewald S, Klöting N, Krause K, Kiess W, Le Duc D, Garten A. Effects of alpelisib treatment on murine Pten-deficient lipomas. Adipocyte 2025; 14:2468275. [PMID: 39962643 PMCID: PMC11844927 DOI: 10.1080/21623945.2025.2468275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/20/2025] [Accepted: 01/31/2025] [Indexed: 02/23/2025] Open
Abstract
Phosphatase and tensin homolog (PTEN) hamartoma tumour syndrome (PHTS) is a rare disorder caused by germline mutations in the tumour suppressor gene PTEN, a key negative regulator of phosphatidylinositol 3-kinase (PI3K)/AKT signalling. Children with PHTS often develop lipomas, for which only surgical resection is available as treatment. We investigated the effects of the selective PI3K-inhibitor alpelisib on Pten-deficient lipomas. After incubation with alpelisib or the non-selective PI3K inhibitor wortmannin, we analysed histology, gene expression, and Pi3k pathway in lipoma and control epididymal adipose tissue (epiWAT). Alpelisib increased adipocyte area in lipomas compared to epiWAT. Baseline gene expression showed higher levels of markers for proliferation (Pcna), fibrosis (Tgfb1), and adipogenesis (Pparg) in lipomas, while hormone-sensitive lipase expression was lower than in epiWAT. Following alpelisib incubation, target genes of Pi3k signalling and extracellular matrix factors were reduced. We confirmed Pi3k inhibition through detecting decreased Akt levels compared to control treatment. Human lipoma samples treated with alpelisib showed variable lipolysis responses, suggesting variability in therapeutic outcomes. We established an ex vivo model to study alpelisib effects on Pten-deficient lipomas. These results underscore the therapeutic potential of targeted PI3K inhibition in the treatment of PHTS-associated lipomas, particularly in cases that are inoperable.
Collapse
Affiliation(s)
- Lea M. Merz
- Center for Pediatric Research, University Hospital for Children & Adolescents, Leipzig University, Leipzig, Germany
| | - Karsten Winter
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Sandy Richter
- Center for Pediatric Research, University Hospital for Children & Adolescents, Leipzig University, Leipzig, Germany
| | - Sonja Kallendrusch
- Institute of Anatomy, Leipzig University, Leipzig, Germany
- Institute of Clinical Research and Systems Medicine, Health and Medical University Potsdam, Potsdam, Germany
| | - Andreas Horn
- Institute of Anatomy, Leipzig University, Leipzig, Germany
| | - Sonja Grunewald
- Department for Dermatology, Venereology and Allergology, University Hospital Leipzig, Leipzig, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG), Helmholtz Center Munich at the University and University Hospital Leipzig, Leipzig, Germany
| | - Kerstin Krause
- Department of Endocrinology, Nephrology and Rheumatology, University Hospital Leipzig, Leipzig, Germany
| | - Wieland Kiess
- Center for Pediatric Research, University Hospital for Children & Adolescents, Leipzig University, Leipzig, Germany
| | - Diana Le Duc
- Institute of Human Genetics, University Hospital Leipzig, Leipzig, Germany
| | - Antje Garten
- Center for Pediatric Research, University Hospital for Children & Adolescents, Leipzig University, Leipzig, Germany
| |
Collapse
|
2
|
Goris M, Jacobsen RG, Lewis AE. Presence of active AKT in the nucleus upon adipocyte differentiation of 3T3-L1 cells. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001140. [PMID: 38495585 PMCID: PMC10940900 DOI: 10.17912/micropub.biology.001140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/14/2024] [Accepted: 02/25/2024] [Indexed: 03/19/2024]
Abstract
AKT is an essential player in the phosphoinositide 3-kinase (PI3K) signalling pathway. Although the mechanisms of its action are well understood at the plasma membrane, AKT can also be found in the nucleus. In adipocytes, this pathway is activated during the process of adipogenesis and solicits both plasma membrane and nuclear AKT activity. However, the endogenous presence of active AKT in the nucleus during adipogenesis has not been shown. Here, we show that the levels of active AKT phosphorylated at Ser-473 increase rapidly after the induction of differentiation in 3T3-L1 cells, both in the cytoplasm and in the nucleus, and tend to remain elevated over the course of differentiation. In conclusion, these results support the notion that nuclear AKT plays an important role in this process.
Collapse
Affiliation(s)
- Marianne Goris
- Department of Biological Sciences, University of Bergen, Bergen, Vestland, Norway
| | - Rhîan G. Jacobsen
- Department of Biological Sciences, University of Bergen, Bergen, Vestland, Norway
| | - Aurélia E. Lewis
- Department of Biological Sciences, University of Bergen, Bergen, Vestland, Norway
| |
Collapse
|
3
|
Lopez-Tello J, Salazar-Petres E, Webb L, Fowden AL, Sferruzzi-Perri AN. Ablation of PI3K-p110alpha Impairs Maternal Metabolic Adaptations to Pregnancy. Front Cell Dev Biol 2022; 10:928210. [PMID: 35846351 PMCID: PMC9283861 DOI: 10.3389/fcell.2022.928210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/10/2022] [Indexed: 01/03/2023] Open
Abstract
Pregnancy requires adaptations in maternal metabolism to support fetal growth. The phosphoinositol-3-kinase (PI3K) signalling pathway controls multiple biological processes and defects in this pathway are linked to metabolic disorders including insulin resistance and glucose intolerance in non-pregnant animals. However, relatively little is known about the contribution of PI3K signalling to the maternal metabolic adaptations during pregnancy. Using mice with partial inactivation of the PI3K isoform, p110α (due to a heterozygous dominant negative mutation; Pik3ca-D933A), the effects of impaired PI3K-p110α signalling on glucose and insulin handling were examined in the pregnant and non-pregnant states and related to the morphological, molecular, and mitochondrial changes in key metabolic organs. The results show that non-pregnant mice lacking PI3K-p110α are glucose intolerant but exhibit compensatory increases in pancreatic glucose-stimulated insulin release and adipose tissue mitochondrial respiratory capacity and fatty acid oxidation. However, in pregnancy, mutant mice failed to show the normal increment in glucose intolerance and pancreatic β-cell mass observed in wild-type pregnant dams and exhibited further enhanced adipose tissue mitochondrial respiratory capacity. These maladaptations in pregnant mutant mice were associated with fetal growth restriction. Hence, PI3K-p110α is a key regulator of metabolic adaptations that support fetal growth during normal pregnancy.
Collapse
Affiliation(s)
| | | | | | | | - Amanda N. Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
4
|
Buchanan CM, Lee KL, Shepherd PR. For Better or Worse: The Potential for Dose Limiting the On-Target Toxicity of PI 3-Kinase Inhibitors. Biomolecules 2019; 9:biom9090402. [PMID: 31443495 PMCID: PMC6770514 DOI: 10.3390/biom9090402] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/15/2019] [Accepted: 08/21/2019] [Indexed: 02/07/2023] Open
Abstract
The hyper-activation of the phosphoinositide (PI) 3-kinase signaling pathway is a hallmark of many cancers and overgrowth syndromes, and as a result, there has been intense interest in the development of drugs that target the various isoforms of PI 3-kinase. Given the key role PI 3-kinases play in many normal cell functions, there is significant potential for the disruption of essential cellular functions by PI 3-kinase inhibitors in normal tissues; so-called on-target drug toxicity. It is, therefore, no surprise that progress within the clinical development of PI 3-kinase inhibitors as single-agent anti-cancer therapies has been slowed by the difficulty of identifying a therapeutic window. The aim of this review is to place the cellular, tissue and whole-body effects of PI 3-kinase inhibition in the context of understanding the potential for dose limiting on-target toxicities and to introduce possible strategies to overcome these.
Collapse
Affiliation(s)
- Christina M Buchanan
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Kate L Lee
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Peter R Shepherd
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|
5
|
Jacobsen RG, Mazloumi Gavgani F, Mellgren G, Lewis AE. DNA Topoisomerase IIα contributes to the early steps of adipogenesis in 3T3-L1 cells. Cell Signal 2016; 28:1593-603. [DOI: 10.1016/j.cellsig.2016.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/07/2016] [Indexed: 01/03/2023]
|
6
|
Smith GC, Ong WK, Costa JL, Watson M, Cornish J, Grey A, Gamble GD, Dickinson M, Leung S, Rewcastle GW, Han W, Shepherd PR. Extended treatment with selective phosphatidylinositol 3-kinase and mTOR inhibitors has effects on metabolism, growth, behaviour and bone strength. FEBS J 2013; 280:5337-49. [PMID: 23837532 DOI: 10.1111/febs.12428] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Revised: 06/24/2013] [Accepted: 06/25/2013] [Indexed: 12/27/2022]
Abstract
The class I phosphatidylinositol 3-kinases (PtdIns3Ks) mediate the effects of many hormones and growth factors on a wide range of cellular processes, and activating mutations or gene amplifications of class I PtdIns3K isoforms are known to contribute to oncogenic processes in a range of tumours. Consequently, a number of small-molecule PtdIns3K inhibitors are under development and in clinical trial. The central signalling role of PtdIns3K in many cellular processes suggests there will be on-target side effects associated with the use of these agents. To gain insights into what these might be we investigated the effect of extended daily dosing of eight small-molecule inhibitors of class Ia PtdIns3Ks. Animals were characterized in metabolic cages to analyse food intake, oxygen consumption and movement. Insulin tolerance and body composition were analysed at the end of the experiment, the latter using EchoMRI. Bone volume and strength was assessed by micro-CT and three-point bending, respectively. Surprisingly, after sustained dosing with pan-PtdIns3K inhibitors and selective inhibitors of the p110α isoform there was a resolution of the impairments in insulin tolerance observed in drug-naïve animals treated with the same drugs. However, pan-PtdIns3K inhibitors and selective inhibitors of the p110α have deleterious effects on animal growth, animal behaviour and bone volume and strength. Together, these findings identify a range of on target effects of PtdIns3K inhibitors and suggest use of these drugs in humans may have important adverse effects on metabolism, body composition, behaviour and skeletal health.
Collapse
Affiliation(s)
- Greg C Smith
- Department of Molecular Medicine and Pathology, University of Auckland, New Zealand
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Ling H, Li X, Yao CH, Hu B, Liao D, Feng S, Wen G, Zhang L. The physiological and pathophysiological roles of adipocyte miRNAs. Biochem Cell Biol 2013; 91:195-202. [PMID: 23859012 DOI: 10.1139/bcb-2012-0053] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
MicroRNAs (miRNAs) are highly conserved, small, noncoding RNAs that regulate gene expression at the posttranscriptional level. Their actions affect numerous important biological processes, including adipocyte differentiation and function, sugar and lipid metabolism, and insulin production and secretion. Recent reports suggest miRNAs may also be involved in the pathogenic processes of obesity, diabetes, and insulin resistance. In this review, we summarize research progresses on adipocyte miRNAs and their physiological and pathological implications.
Collapse
Affiliation(s)
- Hongyan Ling
- Department of Physiology, School of Medicine, University of South China, Hengyang, PR China
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Kim JE, Leung E, Baguley BC, Finlay GJ. Heterogeneity of expression of epithelial-mesenchymal transition markers in melanocytes and melanoma cell lines. Front Genet 2013; 4:97. [PMID: 23755070 PMCID: PMC3668138 DOI: 10.3389/fgene.2013.00097] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Accepted: 05/14/2013] [Indexed: 11/13/2022] Open
Abstract
The epithelial–mesenchymal transition (EMT) describes a reversible switch from an epithelial-like to a mesenchymal-like phenotype. It is essential for the development of the normal epithelium and also contributes to the invasive properties of carcinomas. At the molecular level, the EMT transition is characterized by a series of coordinated changes including downregulation of the junctional protein E-cadherin (CDH1), up-regulation of transcriptional repressors of E-cadherin such as Snail (SNAI1) and Slug (SNAI2), and up-regulation of N-cadherin. We wished to determine whether cultured normal melanocytes and melanoma cell lines, which are derived from the neural crest, showed signs of a similarly coordinated phenotypic switch. We investigated normal melanocytes and 25 cell lines derived from New Zealand patients with metastatic melanoma. Most lines had been previously genotyped for common mutations such as BRAF, NRAS, PIK3CA (phosphatidylinositol-3-kinase), TP53 (p53), and CDKN2A (p16). Expression of E-cadherin, N-cadherin, microphthalmia-associated transcription factor (MITF), Snail, Slug, Axl, p53, and Hdm2 was compared by western blotting. Normal melanocytes expressed each of these proteins except for Snail, while normal melanocytes and almost every melanoma line expressed Slug. Expression of individual markers among different melanoma lines varied from high to low or undetectable. Quantitation of western blots showed that expression of MITF-M, the melanocyte-specific isoform of MITF, was positively related to that of E-cadherin but inversely related to that of N-cadherin and Axl. There was also no apparent relationship between expression of any particular marker and the presence of BRAF, NRAS, PIK3CA, TP53, or CDKN2A mutations. The results suggest that melanomas do not show the classical epithelial and mesenchymal phenotypes but rather display either high E-cadherin/high MITF-M expression on one hand, or high N-cadherin/high Axl expression on the other. These may correspond to differentiated and invasive phenotypes in vivo.
Collapse
Affiliation(s)
- Ji Eun Kim
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland Auckland, New Zealand
| | | | | | | |
Collapse
|
9
|
Foukas LC, Bilanges B, Bettedi L, Pearce W, Ali K, Sancho S, Withers DJ, Vanhaesebroeck B. Long-term p110α PI3K inactivation exerts a beneficial effect on metabolism. EMBO Mol Med 2013; 5:563-71. [PMID: 23483710 PMCID: PMC3628103 DOI: 10.1002/emmm.201201953] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 01/03/2013] [Accepted: 01/07/2013] [Indexed: 11/07/2022] Open
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.
Collapse
Affiliation(s)
- Lazaros C Foukas
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College LondonDarwin Building, Gower Street, London, UK
| | - Benoit Bilanges
- Centre for Cell Signalling, Barts Cancer Institute, Queen Mary University of LondonCharterhouse Square, London, UK
| | - Lucia Bettedi
- Institute of Healthy Ageing and Department of Genetics, Evolution and Environment, University College LondonDarwin Building, Gower Street, London, UK
| | - Wayne Pearce
- Centre for Cell Signalling, Barts Cancer Institute, Queen Mary University of LondonCharterhouse Square, London, UK
| | - Khaled Ali
- Centre for Cell Signalling, Barts Cancer Institute, Queen Mary University of LondonCharterhouse Square, London, UK
| | - Sara Sancho
- Promed Laboratoire MédicalMarly, Switzerland
| | - Dominic J Withers
- Metabolic Signalling Group, Medical Research Council Clinical Sciences Centre, Imperial CollegeDu Cane Road, London, UK
| | - Bart Vanhaesebroeck
- Centre for Cell Signalling, Barts Cancer Institute, Queen Mary University of LondonCharterhouse Square, London, UK
| |
Collapse
|
10
|
Thomas D, Powell JA, Green BD, Barry EF, Ma Y, Woodcock J, Fitter S, Zannettino ACW, Pitson SM, Hughes TP, Lopez AF, Shepherd PR, Wei AH, Ekert PG, Guthridge MA. Protein kinase activity of phosphoinositide 3-kinase regulates cytokine-dependent cell survival. PLoS Biol 2013; 11:e1001515. [PMID: 23526884 PMCID: PMC3601961 DOI: 10.1371/journal.pbio.1001515] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 02/07/2013] [Indexed: 12/20/2022] Open
Abstract
The protein kinase activity of PI3K phosphorylates specific serine residues in growth factor receptors to promote cell survival; these events are constitutively activated in some leukemias. The dual specificity protein/lipid kinase, phosphoinositide 3-kinase (PI3K), promotes growth factor-mediated cell survival and is frequently deregulated in cancer. However, in contrast to canonical lipid-kinase functions, the role of PI3K protein kinase activity in regulating cell survival is unknown. We have employed a novel approach to purify and pharmacologically profile protein kinases from primary human acute myeloid leukemia (AML) cells that phosphorylate serine residues in the cytoplasmic portion of cytokine receptors to promote hemopoietic cell survival. We have isolated a kinase activity that is able to directly phosphorylate Ser585 in the cytoplasmic domain of the interleukin 3 (IL-3) and granulocyte macrophage colony stimulating factor (GM-CSF) receptors and shown it to be PI3K. Physiological concentrations of cytokine in the picomolar range were sufficient for activating the protein kinase activity of PI3K leading to Ser585 phosphorylation and hemopoietic cell survival but did not activate PI3K lipid kinase signaling or promote proliferation. Blockade of PI3K lipid signaling by expression of the pleckstrin homology of Akt1 had no significant impact on the ability of picomolar concentrations of cytokine to promote hemopoietic cell survival. Furthermore, inducible expression of a mutant form of PI3K that is defective in lipid kinase activity but retains protein kinase activity was able to promote Ser585 phosphorylation and hemopoietic cell survival in the absence of cytokine. Blockade of p110α by RNA interference or multiple independent PI3K inhibitors not only blocked Ser585 phosphorylation in cytokine-dependent cells and primary human AML blasts, but also resulted in a block in survival signaling and cell death. Our findings demonstrate a new role for the protein kinase activity of PI3K in phosphorylating the cytoplasmic tail of the GM-CSF and IL-3 receptors to selectively regulate cell survival highlighting the importance of targeting such pathways in cancer. The ability of cells to survive in the absence of proliferation (cell division), differentiation (cell maturation) or activation allows tissues to maintain cell populations that are poised for rapid responses to damage, infections, or other physiological demands. While this “survival-only” response is fundamental to all physiological processes, the underlying mechanisms are not understood. Many growth factors are potent regulators of cell survival through their ability to bind specific cell surface receptors, which in turn activate specialized enzymes called kinases. Phosphoinositide 3-kinase (PI3K) is a dual specificity kinase that is known to be involved in cell survival and malignant transformation, and it is able to phosphorylate both lipid and protein substrates. While the PI3K lipid kinase activity has been extensively studied, the functional significance of its protein kinase activity remains unclear. Here we show that PI3K protein kinase activity can directly phosphorylate growth factor receptors on human hematopoietic (blood) cells to promote a “survival-only” response. We further show that the protein kinase activity of PI3K can be hijacked to result in uncontrolled growth factor receptor phosphorylation and the deregulated survival of leukemic cells. Our studies provide the first evidence that the protein kinase activity of PI3K can control cell survival and that this activity may be deregulated in cancer.
Collapse
Affiliation(s)
- Daniel Thomas
- Cell Growth and Differentiation Laboratory, Division of Human Immunology, Centre for Cancer Biology, SA Pathology, Adelaide, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Kim JE, Stones C, Joseph WR, Leung E, Finlay GJ, Shelling AN, Phillips WA, Shepherd PR, Baguley BC. Comparison of growth factor signalling pathway utilisation in cultured normal melanocytes and melanoma cell lines. BMC Cancer 2012; 12:141. [PMID: 22475322 PMCID: PMC3352269 DOI: 10.1186/1471-2407-12-141] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 04/04/2012] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The phosphatidylinositol-3-kinase (PI3K-PKB), mitogen activated protein kinase (MEK-ERK) and the mammalian target of rapamycin (mTOR- p70S6K), are thought to regulate many aspects of tumour cell proliferation and survival. We have examined the utilisation of these three signalling pathways in a number of cell lines derived from patients with metastatic malignant melanoma of known PIK3CA, PTEN, NRAS and BRAF mutational status. METHODS Western blotting was used to compare the phosphorylation status of components of the PI3K-PKB, MEK-ERK and mTOR-p70S6K signalling pathways, as indices of pathway utilisation. RESULTS Normal melanocytes could not be distinguished from melanoma cells on the basis of pathway utilisation when grown in the presence of serum, but could be distinguished upon serum starvation, where signalling protein phosphorylation was generally abrogated. Surprisingly, the differential utilisation of individual pathways was not consistently associated with the presence of an oncogenic or tumour suppressor mutation of genes in these pathways. CONCLUSION Utilisation of the PI3K-PKB, MEK-ERK and mTOR-p70S6K signalling pathways in melanoma, as determined by phosphorylation of signalling components, varies widely across a series of cell lines, and does not directly reflect mutation of genes coding these components. The main difference between cultured normal melanocytes and melanoma cells is not the pathway utilisation itself, but rather in the serum dependence of pathway utilisation.
Collapse
Affiliation(s)
- Ji Eun Kim
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
- Auckland Cancer Society Research Centre, Private Bag 92019, The University of Auckland, Auckland, New Zealand
| | - Clare Stones
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
| | - Wayne R Joseph
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Euphemia Leung
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
| | - Graeme J Finlay
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
| | - Andrew N Shelling
- Department of Obstetrics and Gynaecology, The University of Auckland, Auckland, New Zealand
| | - Wayne A Phillips
- Department of Surgery, Surgical Oncology Research Laboratory, Peter MacCallum Cancer Centre and University of Melbourne, St. Vincent's Hospital, Melbourne, VIC, Australia
| | - Peter R Shepherd
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| | - Bruce C Baguley
- Auckland Cancer Society Research Centre, The University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
12
|
Abstract
In in vitro studies class-I PI3Ks (phosphoinositide 3-kinases), class-II PI3Ks and mTOR (mammalian target of rapamycin) have all been described as having roles in the regulation of glucose metabolism. The relative role each plays in the normal signalling processes regulating glucose metabolism in vivo is less clear. Knockout and knockin mouse models have provided some evidence that the class-I PI3K isoforms p110α, p110β, and to a lesser extent p110γ, are necessary for processes regulating glucose metabolism and appetite. However, in these models the PI3K activity is chronically reduced. Therefore we analysed the effects of acutely inhibiting PI3K isoforms alone, or PI3K and mTOR, on glucose metabolism and food intake. In the present study impairments in glucose tolerance, insulin tolerance and increased hepatic glucose output were observed in mice treated with the pan-PI3K/mTOR inhibitors PI-103 and NVP-BEZ235. The finding that ZSTK474 has similar effects indicates that these effects are due to inhibition of PI3K rather than mTOR. The p110α-selective inhibitors PIK75 and A66 also induced these phenotypes, but inhibitors of p110β, p110δ or p110γ induced only minor effects. These drugs caused no significant effects on BMR (basal metabolic rate), O2 consumption or water intake, but BEZ235, PI-103 and PIK75 did cause a small reduction in food consumption. Surprisingly, pan-PI3K inhibitors or p110α inhibitors caused reductions in animal movement, although the cause of this is not clear. Taken together these studies provide pharmacological evidence to support a pre-eminent role for the p110α isoform of PI3K in pathways acutely regulating glucose metabolism.
Collapse
|
13
|
Aimaiti A, Wufuer M, Wang YH, Saiyiti M, Cui L, Yusufu A. Can bisphenol A diglycidyl ether (BADGE) administration prevent steroid-induced femoral head osteonecrosis in the early stage? Med Hypotheses 2011; 77:282-5. [PMID: 21620573 DOI: 10.1016/j.mehy.2011.04.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 04/13/2011] [Accepted: 04/28/2011] [Indexed: 12/01/2022]
Abstract
Steroid-induced osteonecrosis of the femoral head (ONFH) is associated with increase of intraosseous pressure caused by elevating of adipogenesis and fat cell hypertrophy in the bone marrow, which subsequently decreases the blood flow in the femoral head and finally resulting in bone ischemia. The early femoral head-preserving method has mainly focused on the conventional core decompression procedure. However, it only achieves a slight decrease in intra-medullary pressure with limited clinical outcome. The crucial point in prevention is to achieve a thorough decompression of intra-medullary pressure and improvement of microcirculation of the femoral head. Bisphenol-A-diglycidyl ether (BADGE), an antagonism of PPAR-γ(Peroxisome proliferator-activated receptor gamma), has been shown to successfully reverse bone marrow adipogenesis and fat cell hypertrophy, enhances proliferation of osteoblasts, inhibit osteoclastogenesis. Therefore, we hypothesized that BADGE administration may be an appropriate novel method for the prevention of early stage steroid-induced ONFH.
Collapse
Affiliation(s)
- Abudusaimi Aimaiti
- Department of Micro-Reconstructive Surgery, The First Teaching Hospital of Xinjiang Medical University, Urumqi 830054, PR China
| | | | | | | | | | | |
Collapse
|
14
|
Moir LM, Trian T, Ge Q, Shepherd PR, Burgess JK, Oliver BGG, Black JL. Phosphatidylinositol 3-kinase isoform-specific effects in airway mesenchymal cell function. J Pharmacol Exp Ther 2011; 337:557-66. [PMID: 21349933 DOI: 10.1124/jpet.110.173583] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K) signal transduction pathway is implicated in the airway remodeling associated with asthma. The class IA PI3K isoforms are known to be activated by growth factors and cytokines. Because this pathway is a possible site of pharmacological intervention for treating the disease, it is important to know which isoforms contribute to this process. Therefore, we used a pharmacological approach to investigate the roles of the three class IA PI3K isoforms (p110α, p110β, and p110δ) in airway remodeling using airway smooth muscle (ASM) cells derived from asthmatic subjects and ASM cells and lung fibroblasts from nonasthmatic subjects. These studies used the inhibitors N'-[(E)-(6-bromoimidazo[1,2-a]pyridin-3-yl)methylidene]-N,2-dimethyl-5-nitrobenzenesulfonohydrazide (PIK75) (which selectively inhibits p110α), 7-methyl-2-(4-morpholinyl)-9-[1-(phenylamino)ethyl]-4H-pyrido[1,2-a]pyrimidin-4-one (TGX221) (which selectively inhibits p110β), and 2-[(6-amino-9H-purin-9-yl)methyl]-5-methyl-3-(2-methylphenyl)-4(3H)-quinazolinone (IC87114) (which selectively inhibits p110δ). Cells were stimulated with transforming growth factor-β (TGFβ) and/or 10% fetal bovine serum in the presence or absence of inhibitor or vehicle control (dimethyl sulfoxide). PIK75, but not TGX221 or IC87114, attenuated TGFβ-induced fibronectin deposition in all cell types tested. PIK75 and TGX221 each decreased secretion of vascular endothelial growth factor and interleukin-6 in nonasthmatic ASM cells and lung fibroblasts, whereas TGX221 was not as effective in asthmatic ASM cells. In addition, PIK75 decreased cell survival in TGFβ-stimulated asthmatic, but not nonasthmatic, ASM cells. In conclusion, specific PI3K isoforms may play a role in pathophysiological events relevant to airway wall remodeling.
Collapse
Affiliation(s)
- Lyn M Moir
- Woolcock Institute of Medical Research, Sydney, Australia.
| | | | | | | | | | | | | |
Collapse
|
15
|
Tups A, Anderson GM, Rizwan M, Augustine RA, Chaussade C, Shepherd PR, Grattan DR. Both p110alpha and p110beta isoforms of phosphatidylinositol 3-OH-kinase are required for insulin signalling in the hypothalamus. J Neuroendocrinol 2010; 22:534-42. [PMID: 20236230 DOI: 10.1111/j.1365-2826.2010.01975.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Both insulin and leptin action in the brain are considered to involve activation of phosphoinositide 3-kinase (PI3K), although the roles of different PI3K isoforms in insulin signalling in the hypothalamus are unknown. In the present study, we characterised the roles of these isoforms in hypothalamic insulin and leptin signalling and investigated the cross-talk of both hormones. To evaluate PI3K levels in the hypothalamus, PI3K was immunoprecipitated using an antibody directed against the p85 subunit, and then total PI3K activity was measured in the presence of novel isoform-selective pharmacological inhibitors of each isoform of PI3K. Subsequently, these inhibitors were administered into the lateral ventricle of male Sprague-Dawley rats, followed by vehicle, insulin, leptin or both hormones 45 min later. PI3K activity was determined by immunohistochemical detection of phosphorylated AKT (S473). In a separate study, the effects of the inhibitors on the anorexigenic action of insulin and leptin were determined. Hypothalamic insulin signalling was specifically mediated by the combined actions of the class Ia isoforms p110alpha and p110beta. Total hypothalamic PI3K activity was inhibited 65% by a p110alpha inhibitor, and 35% by a p110beta inhibitor, with a combination of inhibitors being equally effective as the broad-spectrum PI3K inhibitor wortmannin. Individual i.c.v. administration of p110alpha and p110beta inhibitors partly prevented insulin-induced phosphorylated AKT (S473) in the arcuate nucleus, whereas simultaneous application completely blocked insulin action. Unlike insulin, leptin did not induce phosphorylated AKT in the hypothalamus, as detected by immunohistochemistry, and the anorectic effects of leptin were not affected by pre-treatment with a combination of p110alpha and p110beta inhibitors. The enhanced anorectic effect of a combined i.c.v. application of both insulin and leptin could be prevented by pre-treatment with the combination of p110alpha and p110beta inhibitors. The data suggest that p110alpha and p110beta isoforms of PI3K are necessary to mediate insulin action in the hypothalamus. The role of PI3K in leptin action is less clear, but it may be involved by means of an insulin-dependent sensitisation of leptin action.
Collapse
Affiliation(s)
- A Tups
- Department of Anatomy and Structural Biology, Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand.
| | | | | | | | | | | | | |
Collapse
|
16
|
Grey A, Chaussade C, Empson V, Lin JM, Watson M, O’Sullivan S, Rewcastle G, Naot D, Cornish J, Shepherd P. Evidence for a role for the p110-α isoform of PI3K in skeletal function. Biochem Biophys Res Commun 2010; 391:564-9. [DOI: 10.1016/j.bbrc.2009.11.099] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2009] [Accepted: 11/17/2009] [Indexed: 11/16/2022]
|