Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
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
2.
Biochem Soc Trans ; 51(4): 1459-1472, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37471270

ABSTRACT

The class IA PI3K signaling pathway is activated by growth factor stimulation and regulates a signaling cascade that promotes diverse events including cell growth, proliferation, migration and metabolism. PI3K signaling is one of the most commonly hyperactivated pathways in breast cancer, leading to increased tumor growth and progression. PI3K hyperactivation occurs via a number of genetic and epigenetic mechanisms including mutation or amplification of PIK3CA, the gene encoding the p110α subunit of PI3Kα, as well as via dysregulation of the upstream growth factor receptors or downstream signaling effectors. Over the past decade, extensive efforts to develop therapeutics that suppress oncogenic PI3K signaling have been undertaken. Although FDA-approved PI3K inhibitors are now emerging, their clinical success remains limited due to adverse effects and negative feedback mechanisms which contribute to their reduced efficacy. There is an emerging body of evidence demonstrating crosstalk between the PI3K and Wnt/ß-catenin pathways in breast cancer. However, PI3K exhibits opposing effects on Wnt/ß-catenin signaling in distinct tumor subsets, whereby PI3K promotes Wnt/ß-catenin activation in ER+ cancers, but paradoxically suppresses this pathway in ER- breast cancers. This review discusses the molecular mechanisms for PI3K-Wnt crosstalk in breast cancer, and how Wnt-targeted therapies have the potential to contribute to treatment regimens for breast cancers with PI3K dysregulation.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , beta Catenin/metabolism , Wnt Signaling Pathway/physiology , Cell Proliferation , Cell Line, Tumor
3.
Proc Natl Acad Sci U S A ; 117(45): 28056-28067, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33097662

ABSTRACT

The Rac-GEF, P-Rex1, activates Rac1 signaling downstream of G protein-coupled receptors and PI3K. Increased P-Rex1 expression promotes melanoma progression; however, its role in breast cancer is complex, with differing reports of the effect of its expression on disease outcome. To address this we analyzed human databases, undertook gene array expression analysis, and generated unique murine models of P-Rex1 gain or loss of function. Analysis of PREX1 mRNA expression in breast cancer cDNA arrays and a METABRIC cohort revealed that higher PREX1 mRNA in ER+ve/luminal tumors was associated with poor outcome in luminal B cancers. Prex1 deletion in MMTV-neu or MMTV-PyMT mice reduced Rac1 activation in vivo and improved survival. High level MMTV-driven transgenic PREX1 expression resulted in apicobasal polarity defects and increased mammary epithelial cell proliferation associated with hyperplasia and development of de novo mammary tumors. MMTV-PREX1 expression in MMTV-neu mice increased tumor initiation and enhanced metastasis in vivo, but had no effect on primary tumor growth. Pharmacological inhibition of Rac1 or MEK1/2 reduced P-Rex1-driven tumoroid formation and cell invasion. Therefore, P-Rex1 can act as an oncogene and cooperate with HER2/neu to enhance breast cancer initiation and metastasis, despite having no effect on primary tumor growth.


Subject(s)
Guanine Nucleotide Exchange Factors , Mammary Neoplasms, Experimental , Neoplasm Metastasis , Animals , Cell Polarity/genetics , Female , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Male , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology
4.
Int J Mol Sci ; 24(6)2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36982737

ABSTRACT

Estrogen receptor-positive breast cancers (ER+ BCas) are the most common form of BCa and are increasing in incidence, largely due to changes in reproductive practices in recent decades. Tamoxifen is prescribed as a component of standard-of-care endocrine therapy for the treatment and prevention of ER+ BCa. However, it is poorly tolerated, leading to low uptake of the drug in the preventative setting. Alternative therapies and preventatives for ER+ BCa are needed but development is hampered due to a paucity of syngeneic ER+ preclinical mouse models that allow pre-clinical experimentation in immunocompetent mice. Two ER-positive models, J110 and SSM3, have been reported in addition to other tumour models occasionally shown to express ER (for example 4T1.2, 67NR, EO771, D2.0R and D2A1). Here, we have assessed ER expression and protein levels in seven mouse mammary tumour cell lines and their corresponding tumours, in addition to their cellular composition, tamoxifen sensitivity and molecular phenotype. By immunohistochemical assessment, SSM3 and, to a lesser extent, 67NR cells are ER+. Using flow cytometry and transcript expression we show that SSM3 cells are luminal in nature, whilst D2.0R and J110 cells are stromal/basal. The remainder are also stromal/basal in nature; displaying a stromal or basal Epcam/CD49f FACS phenotype and stromal and basal gene expression signatures are overrepresented in their transcript profile. Consistent with a luminal identity for SSM3 cells, they also show sensitivity to tamoxifen in vitro and in vivo. In conclusion, the data indicate that the SSM3 syngeneic cell line is the only definitively ER+ mouse mammary tumour cell line widely available for pre-clinical research.


Subject(s)
Breast Neoplasms , Receptors, Estrogen , Tamoxifen , Humans , Cell Line, Tumor , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Animals , Mice , Disease Models, Animal , Receptors, Estrogen/genetics , Tamoxifen/pharmacology , Phenotype , Immunohistochemistry , Flow Cytometry , Transcriptome , Mice, 129 Strain , RNA-Seq , Epithelial Cells , Mammary Glands, Animal/cytology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/genetics
5.
Hum Mol Genet ; 29(1): 31-48, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31625572

ABSTRACT

Polycystic kidney disease (PKD) results in the formation of renal cysts that can impair function leading to renal failure. DNA damage accumulates in renal epithelial cells in PKD, but the molecular mechanisms are unclear and are investigated here. Phosphoinositide 3-kinase (PI3K)/AKT signaling activates mammalian target of rapamycin complex 1 (mTORC1) and hyperactivation of mTORC1 is a common event in PKD; however, mTORC1 inhibitors have yielded disappointing results in clinical trials. Here, we demonstrate AKT and mTORC1 hyperactivation in two representative murine PKD models (renal epithelial-specific Inpp5e knockout and collecting duct-specific Pkd1 deletion) and identify a downstream signaling network that contributes to DNA damage accumulation. Inpp5e- and Pkd1-null renal epithelial cells showed DNA damage including double-stranded DNA breaks associated with increased replication fork numbers, multinucleation and centrosome amplification. mTORC1 activated CAD, which promotes de novo pyrimidine synthesis, to sustain cell proliferation. AKT, but not mTORC1, inhibited the DNA repair/replication fork origin firing regulator TOPBP1, which impacts on DNA damage and cell proliferation. Notably, Inpp5e- and Pkd1-null renal epithelial cell spheroid formation defects were rescued by AKT inhibition. These data reveal that AKT hyperactivation contributes to DNA damage accumulation in multiple forms of PKD and cooperates with mTORC1 to promote cell proliferation. Hyperactivation of AKT may play a causal role in PKD by regulating DNA damage and cell proliferation, independent of mTORC1, and AKT inhibition may be a novel therapeutic approach for PKD.


Subject(s)
DNA Damage/physiology , Polycystic Kidney Diseases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Proliferation/genetics , Cell Proliferation/physiology , Cells, Cultured , DNA Damage/genetics , Electrophoresis, Polyacrylamide Gel , Immunoblotting , Immunohistochemistry , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Polycystic Kidney Diseases/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/genetics , Signal Transduction/physiology
6.
Hum Mol Genet ; 28(2): 230-244, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30265301

ABSTRACT

Polycystic kidney disease (PKD) results from excessive renal epithelial cell proliferation, leading to the formation of large fluid filled cysts which impair renal function and frequently lead to renal failure. Hyperactivation of numerous signaling pathways is hypothesized to promote renal epithelial cell hyperproliferation including mTORC1, extracellular signal-regulated kinase (ERK) and WNT signaling. ß-catenin and its target genes are overexpressed in some PKD models and expression of activated ß-catenin induces cysts in mice; however, ß-catenin murine knockout studies indicate it may also inhibit cystogenesis. Therefore, it remains unclear whether ß-catenin is pro- or anti-cystogenic and whether its role is canonical WNT signaling-dependent. Here, we investigate whether ß-catenin deletion in a PKD model with hyperactived ß-catenin signaling affects disease progression to address whether increased ß-catenin drives PKD. We used renal epithelial cell specific Inpp5e-null PKD mice which we report exhibit increased ß-catenin and target gene expression in the cystic kidneys. Surprisingly, co-deletion of ß-catenin with Inpp5e in renal epithelial cells exacerbated polycystic kidney disease and renal failure compared to Inpp5e deletion alone, but did not normalize ß-catenin target gene expression. ß-catenin/Inpp5e double-knockout kidneys exhibited increased cyst initiation, cell proliferation and MEK/ERK signaling compared to Inpp5e-null, associated with increased fibrosis, which may collectively contribute to accelerated disease. Therefore, increased ß-catenin and WNT target gene expression are not necessarily cyst promoting. Rather ß-catenin may play a dual and context-dependent role in PKD and in the presence of other cyst-inducing mutations (Inpp5e-deletion); ß-catenin loss may exacerbate disease in a WNT target gene-independent manner.


Subject(s)
Polycystic Kidney Diseases/metabolism , beta Catenin/metabolism , Animals , Cell Proliferation , Cells, Cultured , Disease Progression , Gene Deletion , Gene Expression , Kidney/metabolism , MAP Kinase Signaling System , Mice , Mice, Knockout , Phosphoric Monoester Hydrolases/genetics , Polycystic Kidney Diseases/enzymology , Polycystic Kidney Diseases/genetics , Signal Transduction , Wnt Proteins/metabolism , beta Catenin/genetics
7.
Int J Mol Sci ; 21(23)2020 Dec 02.
Article in English | MEDLINE | ID: mdl-33276499

ABSTRACT

The phosphoinositide 3-kinase (PI3K)/AKT signalling pathway is hyperactivated in ~70% of breast cancers. Class I PI3K generates PtdIns(3,4,5)P3 at the plasma membrane in response to growth factor stimulation, leading to AKT activation to drive cell proliferation, survival and migration. PTEN negatively regulates PI3K/AKT signalling by dephosphorylating PtdIns(3,4,5)P3 to form PtdIns(4,5)P2. PtdIns(3,4,5)P3 can also be hydrolysed by the inositol polyphosphate 5-phosphatases (5-phosphatases) to produce PtdIns(3,4)P2. Interestingly, while PTEN is a bona fide tumour suppressor and is frequently mutated/lost in breast cancer, 5-phosphatases such as PIPP, SHIP2 and SYNJ2, have demonstrated more diverse roles in regulating mammary tumourigenesis. Reduced PIPP expression is associated with triple negative breast cancers and reduced relapse-free and overall survival. Although PIPP depletion enhances AKT phosphorylation and supports tumour growth, this also inhibits cell migration and metastasis in vivo, in a breast cancer oncogene-driven murine model. Paradoxically, SHIP2 and SYNJ2 are increased in primary breast tumours, which correlates with invasive disease and reduced survival. SHIP2 or SYNJ2 overexpression promotes breast tumourigenesis via AKT-dependent and independent mechanisms. This review will discuss how PTEN, PIPP, SHIP2 and SYNJ2 distinctly regulate multiple functional targets, and the mechanisms by which dysregulation of these distinct phosphoinositide phosphatases differentially affect breast cancer progression.


Subject(s)
Breast Neoplasms/metabolism , Disease Susceptibility , Lipid Metabolism , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Breast Neoplasms/etiology , Breast Neoplasms/pathology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Female , Humans , Inositol Polyphosphate 5-Phosphatases/metabolism , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
8.
J Biol Chem ; 291(33): 17258-70, 2016 08 12.
Article in English | MEDLINE | ID: mdl-27358402

ABSTRACT

PtdIns(3,4,5)P3-dependent Rac exchanger 1 (PREX1) is a Rac-guanine nucleotide exchange factor (GEF) overexpressed in a significant proportion of human breast cancers that integrates signals from upstream ErbB2/3 and CXCR4 membrane surface receptors. However, the PREX1 domains that facilitate its oncogenic activity and downstream signaling are not completely understood. We identify that ERK1/2 MAPK acts downstream of PREX1 and contributes to PREX1-mediated anchorage-independent cell growth. PREX1 overexpression increased but its shRNA knockdown decreased ERK1/2 phosphorylation in response to EGF/IGF-1 stimulation, resulting in induction of the cell cycle regulators cyclin D1 and p21(WAF1/CIP1) PREX1-mediated ERK1/2 phosphorylation, anchorage-independent cell growth, and cell migration were suppressed by inhibition of MEK1/2/ERK1/2 signaling. PREX1 overexpression reduced staurosporine-induced apoptosis whereas its shRNA knockdown promoted apoptosis in response to staurosporine or the anti-estrogen drug tamoxifen. Expression of wild-type but not GEF-inactive PREX1 increased anchorage-independent cell growth. In addition, mouse xenograft studies revealed that expression of wild-type but not GEF-dead PREX1 resulted in the formation of larger tumors that displayed increased phosphorylation of ERK1/2 but not AKT. The impaired anchorage-independent cell growth, apoptosis, and ERK1/2 signaling observed in stable PREX1 knockdown cells was restored by expression of wild-type but not GEF-dead-PREX1. Therefore, PREX1-Rac-GEF activity is critical for PREX1-dependent anchorage-independent cell growth and xenograft tumor growth and may represent a possible therapeutic target for breast cancers that exhibit PREX1 overexpression.


Subject(s)
Breast Neoplasms/metabolism , Cell Proliferation , Gene Expression Regulation, Neoplastic , Guanine Nucleotide Exchange Factors/biosynthesis , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Female , Guanine Nucleotide Exchange Factors/genetics , Humans , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Tamoxifen/pharmacology
9.
Biochem Soc Trans ; 45(4): 963-77, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28710285

ABSTRACT

Phosphatidylinositol 3,4,5-trisphosphate-dependent Rac exchanger (P-Rex) proteins are RacGEFs that are synergistically activated by phosphatidylinositol 3,4,5-trisphosphate and Gßγ subunits of G-protein-coupled receptors. P-Rex1 and P-Rex2 share similar amino acid sequence homology, domain structure, and catalytic function. Recent evidence suggests that both P-Rex proteins may play oncogenic roles in human cancers. P-Rex1 and P-Rex2 are altered predominantly via overexpression and mutation, respectively, in various cancer types, including breast cancer, prostate cancer, and melanoma. This review compares the similarities and differences between P-Rex1 and P-Rex2 functions in human cancers in terms of cellular effects and signalling mechanisms. Emerging clinical data predict that changes in expression or mutation of P-Rex1 and P-Rex2 may lead to changes in tumour outcome, particularly in breast cancer and melanoma.


Subject(s)
Carcinogenesis , Gene Expression Regulation, Neoplastic , Guanine Nucleotide Exchange Factors/metabolism , Models, Molecular , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Animals , Guanine Nucleotide Exchange Factors/agonists , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/genetics , Humans , Mutation , Neoplasm Proteins/agonists , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Neoplasms/genetics , Phosphatidylinositol Phosphates/metabolism , Protein Interaction Domains and Motifs , Protein Subunits/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
10.
Blood ; 125(18): 2815-24, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25736313

ABSTRACT

Phosphoinositide signaling regulates diverse cellular functions. Phosphoinositide-3 kinase (PI3K) generates PtdIns(3,4,5)P3 and PtdIns(3,4)P2, leading to the activation of proliferative and anti-apoptotic signaling pathways. Termination of phosphoinositide signaling requires hydrolysis of inositol ring phosphate groups through the actions of PtdIns(3,4,5)P3 3-phosphatase (PTEN), PtdIns(3,4,5)P3 5-phosphatases (eg, SHIP), and PtdIns(3,4)P2 4-phosphatases (eg, INPP4B). The biological relevance of most of these phosphoinositide phosphatases in acute myeloid leukemia (AML) remains poorly understood. Mass spectrometry-based gene expression profiling of 3-, 4- and 5-phosphatases in human AML revealed significant overexpression of INPP4B. Analysis of an expanded panel of 205 AML cases at diagnosis revealed INPP4B overexpression in association with reduced responses to chemotherapy, early relapse, and poor overall survival, independent of other risk factors. Ectopic overexpression of INPP4B conferred leukemic resistance to cytosine arabinoside (ara-C), daunorubicin, and etoposide. Expression of a phosphatase inert variant (INPP4B C842A) failed to abrogate resistance of AML cells to chemotherapy in vitro or in vivo. In contrast, targeted suppression of endogenously overexpressed INPP4B by RNA interference sensitized AML cell lines and primary AML to chemotherapy. These findings demonstrate a previously unsuspected and clinically relevant role for INPP4B gain of function as a mediator of chemoresistance and poor survival outcome in AML independent of its phosphoinositide phosphatase function.


Subject(s)
Drug Resistance, Neoplasm/genetics , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Phosphoric Monoester Hydrolases/physiology , Adolescent , Adult , Aged , Gene Expression Regulation, Leukemic , Genetic Association Studies , Humans , Leukemia, Myeloid, Acute/mortality , Middle Aged , Phosphoric Monoester Hydrolases/genetics , Polymorphism, Single Nucleotide , Prognosis , Retrospective Studies , Survival Analysis , Transcriptome , Treatment Outcome , Young Adult
11.
J Biol Chem ; 290(34): 20827-20840, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26112412

ABSTRACT

The P-Rex (phosphatidylinositol (3,4,5)-trisphosphate (PIP3)-dependent Rac exchanger) family (P-Rex1 and P-Rex2) of the Rho guanine nucleotide exchange factors (Rho GEFs) activate Rac GTPases to regulate cell migration, invasion, and metastasis in several human cancers. The family is unique among Rho GEFs, as their activity is regulated by the synergistic binding of PIP3 and Gßγ at the plasma membrane. However, the molecular mechanism of this family of multi-domain proteins remains unclear. We report the 1.95 Å crystal structure of the catalytic P-Rex1 DH-PH tandem domain in complex with its cognate GTPase, Rac1 (Ras-related C3 botulinum toxin substrate-1). Mutations in the P-Rex1·Rac1 interface revealed a critical role for this complex in signaling downstream of receptor tyrosine kinases and G protein-coupled receptors. The structural data indicated that the PIP3/Gßγ binding sites are on the opposite surface and markedly removed from the Rac1 interface, supporting a model whereby P-Rex1 binding to PIP3 and/or Gßγ releases inhibitory C-terminal domains to expose the Rac1 binding site.


Subject(s)
Gene Expression Regulation, Neoplastic , Guanine Nucleotide Exchange Factors/chemistry , Phosphatidylinositol Phosphates/chemistry , Recombinant Fusion Proteins/chemistry , rac1 GTP-Binding Protein/chemistry , Amino Acid Sequence , Animals , Baculoviridae/genetics , Catalytic Domain , Crystallography, X-Ray , Enzyme Activation , Female , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , MCF-7 Cells , Models, Molecular , Molecular Sequence Data , Mutation , Phosphatidylinositol Phosphates/metabolism , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sf9 Cells , Signal Transduction , Spodoptera , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism
12.
Prostate ; 75(1): 92-102, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25284366

ABSTRACT

BACKGROUND: Phosphoinositide 3-kinase (PI3K)/Akt pathway is frequently activated in prostate carcinoma due to the loss of tumor suppressor PTEN, which leads to increased Akt activity. Expression of INPP4B, another negative regulator of the PI3K/Akt pathway, is also reduced in prostate carcinoma. However, uncertainty exists regarding the association of INPP4B expression and biochemical and clinical relapse of prostate carcinoma. METHODS: INPP4B expression in benign prostate acini was analyzed by co-immunofluorescence with cytokeratins (CK) 5, 8, 19, androgen receptor (AR), c-MET, chromogranin A and Ki67. INPP4B expression in prostate carcinoma was analyzed in two independent cohorts (n = 406). The association of INPP4B with biochemical and clinical prostate carcinoma relapse was assessed by Kaplan-Meier and Cox proportional hazards modeling. RESULTS: INPP4B was expressed in luminal epithelium within benign ducts, and was highly expressed in CK5+/CK8+/CK19+/AR-/c-MET+/Ki67- intermediate cells in proliferative inflammatory atrophic acini. Overall, INPP4B expression was reduced in prostate carcinoma compared to benign epithelium. Absent/low INPP4B expression was associated with reduced biochemical relapse-free survival (P = 0.01) and increased risk of clinical relapse (P = 0.01). Absence of INPP4B expression was an independent predictor of clinical relapse free survival (P = 0.004) when modeled with Gleason score (P = 0.027) and pathologic stage (P = 0.07). CONCLUSIONS: INPP4B is highly expressed in intermediate cells within proliferative inflammatory atrophic ducts, and expression is reduced in prostate carcinoma. Absence of INPP4B expression is associated with poor outcome following radical prostatectomy, and represents an independent prognostic marker of prostate carcinoma clinical recurrence.


Subject(s)
Chromogranin A/metabolism , Keratins/metabolism , Ki-67 Antigen/metabolism , Phosphoric Monoester Hydrolases/metabolism , Prostatic Neoplasms/enzymology , Proto-Oncogene Proteins c-met/metabolism , Receptors, Androgen/metabolism , Adult , Aged , Disease-Free Survival , Fluorescent Antibody Technique , Fluorescent Antibody Technique, Indirect , Humans , Male , Middle Aged , Neoplasm Recurrence, Local , Proportional Hazards Models , Prostatic Neoplasms/mortality , Prostatic Neoplasms/pathology , Survival Analysis
13.
Blood ; 117(20): 5362-71, 2011 May 19.
Article in English | MEDLINE | ID: mdl-21421839

ABSTRACT

In a recessive ENU mutagenesis screen for embryonic lethality, we identified a mouse pedigree with a missense mutation of SHIP1 (SHIP1(el20)) leading to an amino acid substitution I641T in the inositol-5'-phosphatase domain that represses phosphatidylinositol-3-kinase signaling. Despite detectable expression of functional SHIP1 protein, the phenotype of homozygous SHIP1(el20/el20) mice was more severe than gene-targeted SHIP1-null (SHIP1(-/-)) mice. Compared with age-matched SHIP1(-/-) mice, 5-week-old SHIP1(el20/el20) mice had increased myeloid cells, serum IL-6 levels, marked reductions in lymphoid cells, and died by 7 weeks of age with infiltration of the lungs by activated macrophages. Bone marrow transplantation demonstrated that these defects were hematopoietic-cell-autonomous. We show that the el20 mutation reduces expression in SHIP1(el20/el20) macrophages of both SHIP1 and s-SHIP, an isoform of SHIP1 generated by an internal promoter. In contrast, SHIP1(-/-) macrophages express normal levels of s-SHIP. Compound heterozygous mice (SHIP1(-/el20)) had the same phenotype as SHIP1(-/-) mice, thus providing genetic proof that the more severe phenotype of SHIP1(el20/el20) mice is probably the result of concomitant loss of SHIP1 and s-SHIP. Our results suggest that s-SHIP synergizes with SHIP1 for suppression of macrophage activation, thus providing the first evidence for a role of s-SHIP in adult hematopoiesis.


Subject(s)
Macrophage Activation/genetics , Macrophage Activation/physiology , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/physiology , Amino Acid Substitution , Animals , Base Sequence , Bone Marrow Transplantation , DNA Primers/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Ethylnitrosourea , Female , Genes, Recessive , Hematopoiesis/genetics , Hematopoiesis/physiology , Homozygote , Inositol Polyphosphate 5-Phosphatases , Interleukin-6/biosynthesis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Mutagenesis , Mutation, Missense , Phenotype , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/deficiency , Protein Isoforms/deficiency , Protein Isoforms/genetics , Protein Isoforms/physiology , Signal Transduction
14.
Proc Natl Acad Sci U S A ; 107(51): 22231-6, 2010 Dec 21.
Article in English | MEDLINE | ID: mdl-21127264

ABSTRACT

Inositol polyphosphate 4-phosphatase-II (INPP4B) is a regulator of the phosphoinositide 3-kinase (PI3K) signaling pathway and is implicated as a tumor suppressor in epithelial carcinomas. INPP4B loss of heterozygosity (LOH) is detected in some human breast cancers; however, the expression of INPP4B protein in breast cancer subtypes and the normal breast is unknown. We report here that INPP4B is expressed in nonproliferative estrogen receptor (ER)-positive cells in the normal breast, and in ER-positive, but not negative, breast cancer cell lines. INPP4B knockdown in ER-positive breast cancer cells increased Akt activation, cell proliferation, and xenograft tumor growth. Conversely, reconstitution of INPP4B expression in ER-negative, INPP4B-null human breast cancer cells reduced Akt activation and anchorage-independent growth. INPP4B protein expression was frequently lost in primary human breast carcinomas, associated with high clinical grade and tumor size and loss of hormone receptors and was lost most commonly in aggressive basal-like breast carcinomas. INPP4B protein loss was also frequently observed in phosphatase and tensin homolog (PTEN)-null tumors. These studies provide evidence that INPP4B functions as a tumor suppressor by negatively regulating normal and malignant mammary epithelial cell proliferation through regulation of the PI3K/Akt signaling pathway, and that loss of INPP4B protein is a marker of aggressive basal-like breast carcinomas.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Animals , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Loss of Heterozygosity , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphoric Monoester Hydrolases/genetics , Proto-Oncogene Proteins c-akt/genetics , Transplantation, Heterologous , Tumor Suppressor Proteins/genetics
15.
J Biol Chem ; 286(34): 29758-70, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21712384

ABSTRACT

Phosphoinositide 3-kinase (PI3K) regulates cell polarity and migration by generating phosphatidylinositol 3,4,5-trisphosphate (PI(3,4,5)P(3)) at the leading edge of migrating cells. The serine-threonine protein kinase Akt binds to PI(3,4,5)P(3), resulting in its activation. Active Akt promotes spatially regulated actin cytoskeletal remodeling and thereby directed cell migration. The inositol polyphosphate 5-phosphatases (5-ptases) degrade PI(3,4,5)P(3) to form PI(3,4)P(2), which leads to diminished Akt activation. Several 5-ptases, including SKIP and SHIP2, inhibit actin cytoskeletal reorganization by opposing PI3K/Akt signaling. In this current study, we identify a molecular co-chaperone termed silencer of death domains (SODD/BAG4) that forms a complex with several 5-ptase family members, including SKIP, SHIP1, and SHIP2. The interaction between SODD and SKIP exerts an inhibitory effect on SKIP PI(3,4,5)P(3) 5-ptase catalytic activity and consequently enhances the recruitment of PI(3,4,5)P(3)-effectors to the plasma membrane. In contrast, SODD(-/-) mouse embryonic fibroblasts exhibit reduced Akt-Ser(473) and -Thr(308) phosphorylation following EGF stimulation, associated with increased SKIP PI(3,4,5)P(3)-5-ptase activity. SODD(-/-) mouse embryonic fibroblasts exhibit decreased EGF-stimulated F-actin stress fibers, lamellipodia, and focal adhesion complexity, a phenotype that is rescued by the expression of constitutively active Akt1. Furthermore, reduced cell migration was observed in SODD(-/-) macrophages, which express the three 5-ptases shown to interact with SODD (SKIP, SHIP1, and SHIP2). Therefore, this study identifies SODD as a novel regulator of PI3K/Akt signaling to the actin cytoskeleton.


Subject(s)
Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Stress Fibers/metabolism , Actins/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Movement/physiology , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Focal Adhesions/genetics , Focal Adhesions/metabolism , Inositol Polyphosphate 5-Phosphatases , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/genetics , Proto-Oncogene Proteins c-akt/genetics , Pseudopodia/genetics , Pseudopodia/metabolism , Stress Fibers/genetics
16.
J Biol Chem ; 286(26): 23407-18, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21550974

ABSTRACT

Neuron polarization is essential for the formation of one axon and multiple dendrites, establishing the neuronal circuitry. Phosphoinositide 3-kinase (PI3K) signaling promotes axon selection and elongation. Here we report in hippocampal neurons siRNA knockdown of the proline-rich inositol polyphosphate 5-phosphatase (PIPP), which degrades PI3K-generated PtdIns(3,4,5)P(3), results in multiple hyperelongated axons consistent with a polarization defect. We identify collapsin response mediator protein 2 (CRMP2), which regulates axon selection by promoting WAVE1 delivery via Kinesin-1 motors to the axon growth cone, as a PIPP-interacting protein by Y2H screening, direct binding studies, and coimmunoprecipitation of an endogenous PIPP, CRMP2, and Kinesin-1 complex from brain lysates. The C-terminal growth cone-targeting domain of PIPP facilitates its interaction with CRMP2. PIPP growth cone localization is CRMP2-dependent. PIPP knockdown in PC12 cells promotes neurite elongation, WAVE1 and Kinesin-1 growth cone localization, whereas knockdown of CRMP2 exhibits the opposite phenotype, with shorter neurites and decreased WAVE1/Kinesin-1 at the growth cone. In contrast, CRMP2 overexpression promotes neurite elongation, a phenotype rescued by full-length PIPP, or expression of the CRMP2-binding PIPP domain. Therefore this study identifies PIPP and CRMP2 exert opposing roles in promoting axon selection and neurite elongation and the complex between these proteins serves to regulate the localization of effectors that promote neurite extension.


Subject(s)
Growth Cones/metabolism , Hippocampus/metabolism , Multiprotein Complexes/metabolism , Nerve Tissue Proteins/metabolism , Neurites/metabolism , Phosphoric Monoester Hydrolases/metabolism , Animals , Hippocampus/cytology , Inositol Polyphosphate 5-Phosphatases , Intercellular Signaling Peptides and Proteins , Kinesins/genetics , Kinesins/metabolism , Male , Multiprotein Complexes/genetics , Nerve Tissue Proteins/genetics , PC12 Cells , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol Phosphates/genetics , Phosphatidylinositol Phosphates/metabolism , Phosphoric Monoester Hydrolases/genetics , Rats , Rats, Sprague-Dawley , Wiskott-Aldrich Syndrome Protein Family/genetics , Wiskott-Aldrich Syndrome Protein Family/metabolism
17.
Cancers (Basel) ; 15(1)2022 Dec 26.
Article in English | MEDLINE | ID: mdl-36612130

ABSTRACT

The majority of breast cancers are estrogen receptor-positive (ER+), and endocrine therapies that suppress ER signaling are the standard-of-care treatment for this subset. However, up to half of all ER+ cancers eventually relapse, highlighting a need for improved clinical therapies. The phosphoinositide phosphatase, INPP4B, is overexpressed in almost half of all ER+ breast cancers, and promotes Wnt/ß-catenin signaling, cell proliferation and tumor growth. Here, using cell viability assays, we report that INPP4B overexpression does not affect the sensitivity of ER+ breast cancer cells to standard-of-care treatments including the anti-estrogen 4-hydroxytamoxifen (4-OHT) or the PI3Kα inhibitor alpelisib. Examination of four small molecule Wnt inhibitors revealed that ER+ breast cancer cells with INPP4B overexpression were more sensitive to the FDA-approved drug pyrvinium and a 4-OHT-pyrvinium combination treatment. Using 3D culture models, we demonstrated that pyrvinium selectively reduced the size of INPP4B-overexpressing ER+ breast cancer spheroids in the presence and absence of 4-OHT. These findings suggest that repurposing pyrvinium as a Wnt inhibitor may be an effective therapeutic strategy for human ER+ breast cancers with high INPP4B levels.

18.
Mol Cell Oncol ; 8(4): 1954470, 2021.
Article in English | MEDLINE | ID: mdl-34616876

ABSTRACT

AKT is the central phosphoinositide 3-kinase (PI3K) signaling effector, however, PIK3CA (p110α subunit of PI3Kα)-mutant estrogen receptor-positive (ER+) breast cancers exhibit minimal AKT activation and the downstream signaling is poorly characterized. We discovered that a subset of PIK3CA-mutant ER+ breast cancers exhibit increased inositol polyphosphate 4-phosphatase type II (INPP4B) expression, which promotes late endosome formation and glycogen synthase kinase 3 beta (GSK3ß) trafficking, leading to enhanced Wingless-related integration site (WNT)/catenin beta 1 (ß-catenin) activation.

19.
Adv Biol Regul ; 82: 100817, 2021 12.
Article in English | MEDLINE | ID: mdl-34216856

ABSTRACT

Cancer is a complex and heterogeneous disease marked by the dysregulation of cancer driver genes historically classified as oncogenes or tumour suppressors according to their ability to promote or inhibit tumour development and growth, respectively. Certain genes display both oncogenic and tumour suppressor functions depending on the biological context, and as such have been termed dual-role cancer driver genes. However, because of their context-dependent behaviour, the tumourigenic mechanism of many dual-role genes is elusive and remains a significant knowledge gap in our effort to understand and treat cancer. Inositol polyphosphate 4-phosphatase type II (INPP4B) is an emerging dual-role cancer driver gene, primarily known for its role as a negative regulator of the phosphoinositide 3-kinase (PI3K)/AKT signalling pathway. In response to growth factor stimulation, class I PI3K generates PtdIns(3,4,5)P3 at the plasma membrane. PtdIns(3,4,5)P3 can be hydrolysed by inositol polyphosphate 5-phosphatases to generate PtdIns(3,4)P2, which, together with PtdIns(3,4,5)P3, facilitates the activation of AKT to promote cell proliferation, survival, migration, and metabolism. Phosphatase and tensin homology on chromosome 10 (PTEN) and INPP4B are dual-specificity phosphatases that hydrolyse PtdIns(3,4,5)P3 and PtdIns(3,4)P2, respectively, and thus negatively regulate PI3K/AKT signalling. PTEN is a bona fide tumour suppressor that is frequently lost in human tumours. INPP4B was initially characterised as a tumour suppressor akin to PTEN, and has been implicated as such in a number of cancers, including prostate, thyroid, and basal-like breast cancers. However, evidence has since emerged revealing INPP4B as a paradoxical oncogene in several malignancies, with increased INPP4B expression reported in AML, melanoma and colon cancers among others. Although the tumour suppressive function of INPP4B has been mostly ascribed to its ability to negatively regulate PI3K/AKT signalling, its oncogenic function remains less clear, with proposed mechanisms including promotion of PtdIns(3)P-dependent SGK3 signalling, inhibition of PTEN-dependent AKT activation, and enhancing DNA repair mechanisms to confer chemoresistance. Nevertheless, research is ongoing to identify the factors that dictate the tumourigenic output of INPP4B in different human cancers. In this review we discuss the dualistic role that INPP4B plays in the context of cancer development, progression and treatment, drawing comparisons to PTEN to explore how their similarities and, importantly, their differences may account for their diverging roles in tumourigenesis.


Subject(s)
Neoplasms/genetics , Phosphoric Monoester Hydrolases , Genes, Tumor Suppressor , Humans , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
20.
Nat Commun ; 12(1): 3140, 2021 05 25.
Article in English | MEDLINE | ID: mdl-34035258

ABSTRACT

INPP4B suppresses PI3K/AKT signaling by converting PI(3,4)P2 to PI(3)P and INPP4B inactivation is common in triple-negative breast cancer. Paradoxically, INPP4B is also a reported oncogene in other cancers. How these opposing INPP4B roles relate to PI3K regulation is unclear. We report PIK3CA-mutant ER+ breast cancers exhibit increased INPP4B mRNA and protein expression and INPP4B increased the proliferation and tumor growth of PIK3CA-mutant ER+ breast cancer cells, despite suppression of AKT signaling. We used integrated proteomics, transcriptomics and imaging to demonstrate INPP4B localized to late endosomes via interaction with Rab7, which increased endosomal PI3Kα-dependent PI(3,4)P2 to PI(3)P conversion, late endosome/lysosome number and cargo trafficking, resulting in enhanced GSK3ß lysosomal degradation and activation of Wnt/ß-catenin signaling. Mechanistically, Wnt inhibition or depletion of the PI(3)P-effector, Hrs, reduced INPP4B-mediated cell proliferation and tumor growth. Therefore, INPP4B facilitates PI3Kα crosstalk with Wnt signaling in ER+ breast cancer via PI(3,4)P2 to PI(3)P conversion on late endosomes, suggesting these tumors may be targeted with combined PI3K and Wnt/ß-catenin therapies.


Subject(s)
Breast Neoplasms/pathology , Class I Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Carcinogenesis/drug effects , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Class I Phosphatidylinositol 3-Kinases/genetics , Endosomes/metabolism , Female , Gene Expression Profiling , Humans , Lysosomes/metabolism , Mice , Mutation , Phosphatidylinositol Phosphates/metabolism , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Proteolysis/drug effects , Proteomics , Thiazoles/pharmacology , Thiazoles/therapeutic use , Tissue Array Analysis , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL