Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
Eur J Nucl Med Mol Imaging ; 50(9): 2621-2635, 2023 07.
Article in English | MEDLINE | ID: mdl-37086273

ABSTRACT

PURPOSE: FAP is a membrane-bound protease under investigation as a pan-cancer target, given its high levels in tumors but limited expression in normal tissues. FAP-2286 is a radiopharmaceutical in clinical development for solid tumors that consists of two functional elements: a FAP-targeting peptide and a chelator used to attach radioisotopes. Preclinically, we evaluated the immune modulation and anti-tumor efficacy of FAP-2287, a murine surrogate for FAP-2286, conjugated to the radionuclide lutetium-177 (177Lu) as a monotherapy and in combination with a PD-1 targeting antibody. METHODS: C57BL/6 mice bearing MCA205 mouse FAP-expressing tumors (MCA205-mFAP) were treated with 177Lu-FAP-2287, anti-PD-1, or both. Tumor uptake of 177Lu- FAP-2287 was assessed by SPECT/CT scanning, while therapeutic efficacy was measured by tumor volume and survival. Immune profiling of tumor infiltrates was evaluated through flow cytometry, RNA expression, and immunohistochemistry analyses. RESULTS: 177Lu-FAP-2287 rapidly accumulated in MCA205-mFAP tumors leading to significant tumor growth inhibition (TGI) and longer survival time. Significant TGI was also observed from anti-PD-1 and the combination. In flow cytometry analysis of tumors, 177Lu-FAP-2287 increased CD8+ T cell infiltration which was maintained in the combination with anti-PD-1. The increase in CD8+ T cells was accompanied by an induction of STING-mediated type I interferon response and higher levels of co-stimulatory molecules such as CD86. CONCLUSION: In a preclinical model, FAP-targeted radiotherapy enhanced anti-PD-1-mediated TGI by modulating the TME and increasing the recruitment of tumor-infiltrating CD8+ T cells. These findings provide a rationale for clinical studies of combined 177Lu-FAP-2286 radiotherapy and immune checkpoint inhibition in FAP-positive tumors.


Subject(s)
CD8-Positive T-Lymphocytes , Immune Checkpoint Inhibitors , Animals , Mice , Tumor Microenvironment , Cell Line, Tumor , Mice, Inbred C57BL , Fibroblasts
2.
J Immunother ; 45(8): 335-348, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35791438

ABSTRACT

Lucitanib is a multi-tyrosine kinase inhibitor whose targets are associated with angiogenesis and other key cancer and immune pathways. Its antiangiogenic properties are understood, but lucitanib's immunomodulatory activity is heretofore unknown. Lucitanib exhibited such activity in vivo, increasing CD3 + , CD8 + , and CD4 + T cells and decreasing dendritic cells and monocyte-derived suppressor cells in mouse spleens. Depletion of CD8 + T cells from syngeneic MC38 colon tumor-bearing mice reduced the antitumor efficacy of lucitanib and revealed a CD8 + T-cell-dependent component of lucitanib's activity. The combination of lucitanib and costimulatory immune pathway agonists targeting 4-1BB, glucocorticoid-induced TNFR (GITR), inducible T-cell co-stimulator (ICOS), or OX40 exhibited enhanced antitumor activity compared with each single agent in immunocompetent tumor models. Lucitanib combined with blockade of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) or programmed cell death protein-1 (PD-1) coinhibitory immune pathways also showed enhanced antitumor activity over the single agents in multiple models. In CT26 tumors, lucitanib, alone or combined with anti-PD-1, reduced CD31 + vessels and depleted F4/80 + macrophages. Combination treatment also increased the number of intratumoral T cells. Gene expression in pathways associated with immune activity was upregulated by lucitanib in MC38 tumors and further potentiated by combination with anti-PD-1. Accordingly, lucitanib, alone or combined with anti-PD-1, increased intratumoral CD8 + T-cell abundance. Lucitanib's antitumor and pharmacodynamic activity, alone or combined with anti-PD-1, was not recapitulated by specific vascular endothelial growth factor receptor-2 (VEGFR2) inhibition. These data indicate that lucitanib can modulate vascular and immune components of the tumor microenvironment and cooperate with immunotherapy to enhance antitumor efficacy. They support the clinical development of lucitanib combined with immune pathway modulators to treat cancer.


Subject(s)
Antineoplastic Agents , Neoplasms , Quinolines , Animals , Antineoplastic Agents/therapeutic use , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Immunologic Factors/therapeutic use , Mice , Naphthalenes , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Quinolines/pharmacology , Quinolines/therapeutic use , Tumor Microenvironment
3.
Nat Med ; 25(1): 111-118, 2019 01.
Article in English | MEDLINE | ID: mdl-30478424

ABSTRACT

Although targeted therapies often elicit profound initial patient responses, these effects are transient due to residual disease leading to acquired resistance. How tumors transition between drug responsiveness, tolerance and resistance, especially in the absence of preexisting subclones, remains unclear. In epidermal growth factor receptor (EGFR)-mutant lung adenocarcinoma cells, we demonstrate that residual disease and acquired resistance in response to EGFR inhibitors requires Aurora kinase A (AURKA) activity. Nongenetic resistance through the activation of AURKA by its coactivator TPX2 emerges in response to chronic EGFR inhibition where it mitigates drug-induced apoptosis. Aurora kinase inhibitors suppress this adaptive survival program, increasing the magnitude and duration of EGFR inhibitor response in preclinical models. Treatment-induced activation of AURKA is associated with resistance to EGFR inhibitors in vitro, in vivo and in most individuals with EGFR-mutant lung adenocarcinoma. These findings delineate a molecular path whereby drug resistance emerges from drug-tolerant cells and unveils a synthetic lethal strategy for enhancing responses to EGFR inhibitors by suppressing AURKA-driven residual disease and acquired resistance.


Subject(s)
Aurora Kinase A/metabolism , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Protein Kinase Inhibitors/therapeutic use , Animals , Apoptosis/drug effects , Cell Count , Cell Cycle Proteins/metabolism , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Mice , Microtubule-Associated Proteins/metabolism , Mutation/genetics , Neoplasm, Residual/drug therapy , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology
4.
Cancer Discov ; 7(9): 984-998, 2017 09.
Article in English | MEDLINE | ID: mdl-28588062

ABSTRACT

High-grade epithelial ovarian carcinomas containing mutated BRCA1 or BRCA2 (BRCA1/2) homologous recombination (HR) genes are sensitive to platinum-based chemotherapy and PARP inhibitors (PARPi), while restoration of HR function due to secondary mutations in BRCA1/2 has been recognized as an important resistance mechanism. We sequenced core HR pathway genes in 12 pairs of pretreatment and postprogression tumor biopsy samples collected from patients in ARIEL2 Part 1, a phase II study of the PARPi rucaparib as treatment for platinum-sensitive, relapsed ovarian carcinoma. In 6 of 12 pretreatment biopsies, a truncation mutation in BRCA1, RAD51C, or RAD51D was identified. In five of six paired postprogression biopsies, one or more secondary mutations restored the open reading frame. Four distinct secondary mutations and spatial heterogeneity were observed for RAD51CIn vitro complementation assays and a patient-derived xenograft, as well as predictive molecular modeling, confirmed that resistance to rucaparib was associated with secondary mutations.Significance: Analyses of primary and secondary mutations in RAD51C and RAD51D provide evidence for these primary mutations in conferring PARPi sensitivity and secondary mutations as a mechanism of acquired PARPi resistance. PARPi resistance due to secondary mutations underpins the need for early delivery of PARPi therapy and for combination strategies. Cancer Discov; 7(9); 984-98. ©2017 AACR.See related commentary by Domchek, p. 937See related article by Quigley et al., p. 999See related article by Goodall et al., p. 1006This article is highlighted in the In This Issue feature, p. 920.


Subject(s)
DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm/genetics , Indoles/therapeutic use , Ovarian Neoplasms/drug therapy , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Animals , CHO Cells , Cell Line, Tumor , Cricetulus , Female , HEK293 Cells , Humans , Mutation , Ovarian Neoplasms/genetics
5.
Proteomics ; 16(14): 1992-7, 2016 07.
Article in English | MEDLINE | ID: mdl-27282143

ABSTRACT

The PI3K pathway is commonly activated in cancer. Only a few studies have attempted to explore the spectrum of phosphorylation signaling downstream of the PI3K cascade. Such insight, however, is imperative to understand the mechanisms responsible for oncogenic phenotypes. By applying MS-based phosphoproteomics, we mapped 2509 phosphorylation sites on 1096 proteins, and quantified their responses to activation or inhibition of PIK3CA using isogenic knock-in derivatives and a series of targeted inhibitors. We uncovered phosphorylation changes in a wide variety of proteins involved in cell growth and proliferation, many of which have not been previously associated with PI3K signaling. A significant update of the posttranslational modification database PHOSIDA (http://www.phosida.com) allows efficient use of the data. All MS data have been deposited in the ProteomeXchange with identifier PXD003899 (http://proteomecentral.proteomexchange.org/dataset/PXD003899).


Subject(s)
Cell Transformation, Neoplastic/genetics , Epithelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphoproteins/genetics , Protein Processing, Post-Translational , Antineoplastic Agents/pharmacology , Cell Line , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Class I Phosphatidylinositol 3-Kinases , Colon/cytology , Colon/drug effects , Colon/metabolism , Databases, Genetic , Epithelial Cells/cytology , Epithelial Cells/drug effects , Humans , Internet , Mutation , Neoplasm Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proteomics/methods , Signal Transduction , Software
6.
Neoplasia ; 15(7): 694-711, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23814482

ABSTRACT

The phosphatidylinositol 3-kinase (PI3K) pathway is a central mediator of vascular endothelial growth factor (VEGF)-driven angiogenesis. The discovery of small molecule inhibitors that selectively target PI3K or PI3K and mammalian target of rapamycin (mTOR) provides an opportunity to pharmacologically determine the contribution of these key signaling nodes in VEGF-A-driven tumor angiogenesis in vivo. This study used an array of micro-vascular imaging techniques to monitor the antivascular effects of selective class I PI3K, mTOR, or dual PI3K/mTOR inhibitors in colorectal and prostate cancer xenograft models. Micro-computed tomography (micro-CT) angiography, dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), vessel size index (VSI) MRI, and DCE ultrasound (DCE-U/S) were employed to quantitatively evaluate the vascular (structural and physiological) response to these inhibitors. GDC-0980, a dual PI3K/mTOR inhibitor, was found to reduce micro-CT angiography vascular density, while VSI MRI demonstrated a significant reduction in vessel density and an increase in mean vessel size, consistent with a loss of small functional vessels and a substantial antivascular response. DCE-MRI showed that GDC-0980 produces a strong functional response by decreasing the vascular permeability/perfusion-related parameter, K (trans). Interestingly, comparable antivascular effects were observed for both GDC-980 and GNE-490 (a selective class I PI3K inhibitor). In addition, mTOR-selective inhibitors did not affect vascular density, suggesting that PI3K inhibition is sufficient to generate structural changes, characteristic of a robust antivascular response. This study supports the use of noninvasive microvascular imaging techniques (DCE-MRI, VSI MRI, DCE-U/S) as pharmacodynamic assays to quantitatively measure the activity of PI3K and dual PI3K/mTOR inhibitors in vivo.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/antagonists & inhibitors , Enzyme Inhibitors , Neoplasms/diagnosis , Neovascularization, Pathologic/diagnosis , Angiography/methods , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Class I Phosphatidylinositol 3-Kinases/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Heterografts , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Magnetic Resonance Imaging/methods , Mice , Multimodal Imaging , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Pyrimidines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tumor Burden/drug effects , Ultrasonography/methods , X-Ray Microtomography/methods
7.
Cancer Lett ; 326(2): 168-75, 2012 Dec 30.
Article in English | MEDLINE | ID: mdl-22902509

ABSTRACT

The mammalian target of rapamycin (mTOR) is a central node in a complex signaling network that is regulated by several pathways deregulated in human cancers, including the PI3K/Akt and MAPK pathways. Targeting mTOR therefore presents an opportunity for therapeutic intervention. However, mTOR inhibition with rapamycin analogs or kinase inhibitors reduces cell growth but does not induce apoptosis, and the clinical benefit of rapamycin analogs has been modest. In this study we show that mTOR kinase inhibitors can potentiate apoptosis when used in combination with upstream targeted agents such as PI3K and MEK inhibitors. This increased apoptosis is dependent on genetic background, and correlates with active growth factor survival pathways. In PI3K mutant tumors, mTOR inhibition leads to partial reactivation of Akt which allows cells to survive, whereas in KRAS mutant tumors, this same reactivation of Akt occurs but is not required for cell survival. These data suggest the use of selected rational combinations of mTOR kinase inhibitors with other targeted inhibitors in specific tumor genotypes to achieve the maximal cytotoxic response by inhibiting two nodes in the activated signaling network.


Subject(s)
Apoptosis/drug effects , MAP Kinase Kinase Kinases/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Blotting, Western , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Humans
8.
Eur J Pharmacol ; 577(1-3): 44-53, 2007 Dec 22.
Article in English | MEDLINE | ID: mdl-17869243

ABSTRACT

Altered regulation of dopamine D(2) receptors is implicated in addiction, schizophrenia and movement disorders, as well as lactotroph growth and regulation. Dopamine D(2S) and dopamine D(2L) receptors are alternately-spliced variants that differ by 29 amino acids in the third intracellular (i3) domain and display different sensitivity to desensitization by protein kinase C (PKC). In the present studies we determined the specific phosphorylation sites on the dopamine D(2S) receptor that confer PKC-mediated desensitization. In dopamine D(2L) receptors, we identified a PKC pseudosubstrate site responsible for the relative insensitivity of the receptor to PKC-induced uncoupling. In transiently transfected Ltk(-) fibroblast cells, 2-min preactivation of PKC with 12-O-tetradecanoyl 4beta-phorbol 13alpha-acetate (TPA) completely inhibited calcium mobilization induced by the dopamine D(2S) receptor, but not the dopamine D(2L) variant. Point mutation of i3 PKC sites Ser228/229Gly rendered the dopamine D(2S) receptor resistant to PKC action, with lesser effects of other Ser and Thr mutations. Inactivation of the PKC pseudosubstrate motif in the dopamine D(2L) receptor sensitized the receptor to PKC, and this was reversed by mutation of i3 PKC sites Ser228/229. A phospho-specific antibody generated against phospho-Ser228/229 demonstrated PKC-induced phosphorylation at these sites of dopamine D(2S), but not D(2L) receptors, in Ltk(-) cells. Conversely, the pseudosubstrate dopamine D(2L) receptor mutant displayed PKC-induced phosphorylation at Ser228/229, which was abolished when these sites were mutated. Similar phosphorylation results were observed using GH4 cells stably transfected with dopamine D(2) receptors and mutants. Thus the relative location of phosphorylation and pseudosubstrate sites provides an important determinant substrate sensitivity to PKC.


Subject(s)
Dopamine D2 Receptor Antagonists , Protein Kinase C/pharmacology , Animals , Antibodies, Blocking/pharmacology , Binding Sites , Blotting, Western , Calcium/metabolism , Cells, Cultured , Ligands , Mice , Mutagenesis/physiology , Mutation/physiology , Phosphorylation , Pituitary Gland/cytology , Pituitary Gland/metabolism , Receptors, Dopamine D2/genetics , Tetradecanoylphorbol Acetate/pharmacology , Transfection
9.
J Biol Chem ; 282(6): 3929-39, 2007 Feb 09.
Article in English | MEDLINE | ID: mdl-17145753

ABSTRACT

Up-regulation of urokinase receptors is common during tumor progression and thought to promote invasion and metastasis. Urokinase receptors bind urokinase and a set of beta1 integrins, but it remains unclear to what degree urokinase receptor/integrin binding is important to beta1 integrin signaling. Using site-directed mutagenesis, single amino acid mutants of the urokinase receptor were identified that fail to associate with either alpha3beta1 (D262A) or alpha5beta1 (H249A) but associate normally with urokinase. To study the effects of these mutations on beta1 integrin function, endogenous urokinase receptors were first stably silenced in tumor cell lines HT1080 and H1299, and then wild type or mutant receptors were expressed. Knockdown of urokinase receptors resulted in markedly reduced fibronectin and alpha5beta1-dependent ERK activation and metalloproteinase MMP-9 expression. Re-expression of wild type or D262A mutant receptors but not the alpha5beta1 binding-deficient H249A mutant reconstituted fibronectin responses. Because urokinase receptor.alpha5beta1 complexes bind in the fibronectin heparin-binding domain (Type III 12-14) whereas alpha5beta1 primarily binds in the RGD-containing domain (Type III 7-10), signaling pathways leading to ERK and MMP-9 responses were dissected. Binding to III 7-10 led to Src/focal adhesion kinase activation, whereas binding to III 7-14 caused Rac 1 activation. Tumor cells engaging fibronectin required both Type III 7-10- and 12-14-initiated signals to activate ERK and up-regulate MMP-9. Thus urokinase receptor binding to alpha5beta1 is required for maximal responses to fibronectin and tumor cell invasion, and this operates through an enhanced Src/Rac/ERK signaling pathway.


Subject(s)
Fibrosarcoma/enzymology , Integrin alpha5beta1/physiology , Lung Neoplasms/enzymology , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Urokinase-Type Plasminogen Activator/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Fibronectins/metabolism , Fibronectins/physiology , Fibrosarcoma/genetics , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Gene Deletion , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Protein Binding/genetics , Protein Binding/physiology , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Urokinase Plasminogen Activator , Signal Transduction/genetics
10.
Proc Natl Acad Sci U S A ; 103(35): 13180-5, 2006 Aug 29.
Article in English | MEDLINE | ID: mdl-16924102

ABSTRACT

Mechanisms leading to fibroblast accumulation during pulmonary fibrogenesis remain unclear. Although there is in vitro evidence of lung alveolar epithelial-to-mesenchymal transition (EMT), whether EMT occurs within the lung is currently unknown. Biopsies from fibrotic human lungs demonstrate epithelial cells with mesenchymal features, suggesting EMT. To more definitively test the capacity of alveolar epithelial cells for EMT, mice expressing beta-galactosidase (beta-gal) exclusively in lung epithelial cells were generated, and their fates were followed in an established model of pulmonary fibrosis, overexpression of active TGF-beta1. beta-gal-positive cells expressing mesenchymal markers accumulated within 3 weeks of in vivo TGF-beta1 expression. The increase in vimentin-positive cells within injured lungs was nearly all beta-gal-positive, indicating epithelial cells as the main source of mesenchymal expansion in this model. Ex vivo, primary alveolar epithelial cells cultured on provisional matrix components, fibronectin or fibrin, undergo robust EMT via integrin-dependent activation of endogenous latent TGF-beta1. In contrast, primary cells cultured on laminin/collagen mixtures do not activate the TGF-beta1 pathway and, if exposed to active TGF-beta1, undergo apoptosis rather than EMT. These data reveal alveolar epithelial cells as progenitors for fibroblasts in vivo and implicate the provisional extracellular matrix as a key regulator of epithelial transdifferentiation during fibrogenesis.


Subject(s)
Epithelial Cells/cytology , Extracellular Matrix/metabolism , Mesoderm/cytology , Pulmonary Alveoli/cytology , Pulmonary Fibrosis/pathology , Animals , Apoptosis , Cells, Cultured , Collagen/metabolism , Drug Combinations , Fibronectins/metabolism , Genes, Reporter , Humans , Laminin/metabolism , Mice , Mice, Transgenic , Proteoglycans/metabolism , Pulmonary Alveoli/pathology , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , beta-Galactosidase/metabolism
11.
Mol Cell Biol ; 26(6): 2309-16, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16508006

ABSTRACT

Cathepsin F (cat F) is a widely expressed lysosomal cysteine protease whose in vivo role is unknown. To address this issue, mice deficient in cat F were generated via homologous recombination. Although cat F-/- mice appeared healthy and reproduced normally, they developed progressive hind leg weakness and decline in motor coordination at 12 to 16 months of age, followed by significant weight loss and death within 6 months. cat F was found to be expressed throughout the central nervous system (CNS). cat F-/- neurons accumulated eosinophilic granules that had features typical of lysosomal lipofuscin by electron microscopy. Large amounts of autofluorescent lipofuscin, characteristic of the neurodegenerative disease neuronal ceroid lipofuscinosis (NCL), accumulated throughout the CNS but not in visceral organs, beginning as early as 6 weeks of age. Pronounced gliosis, an indicator of neuronal stress and neurodegeneration, was also apparent in older cat F-/- mice. cat F is the only cysteine cathepsin whose inactivation alone causes a lysosomal storage defect and progressive neurological features in mice. The late onset suggests that this gene may be a candidate for adult-onset NCL.


Subject(s)
Cathepsins/deficiency , Nervous System Diseases/etiology , Neuronal Ceroid-Lipofuscinoses/etiology , Age Factors , Age of Onset , Animals , Cathepsin F , Cathepsins/genetics , Cathepsins/metabolism , Central Nervous System/metabolism , Central Nervous System/pathology , Lipofuscin/metabolism , Mice , Mice, Mutant Strains , Motor Neurons/pathology , Motor Neurons/ultrastructure , Neuromuscular Diseases/etiology , Neuronal Ceroid-Lipofuscinoses/epidemiology , Neuronal Ceroid-Lipofuscinoses/pathology , Neurons/pathology , Sequence Analysis, DNA , Spinal Cord/pathology , Spinal Cord/ultrastructure , Weight Loss
12.
J Cell Biol ; 168(3): 501-11, 2005 Jan 31.
Article in English | MEDLINE | ID: mdl-15684035

ABSTRACT

Urokinase-type plasminogen activator receptors (uPARs), up-regulated during tumor progression, associate with beta1 integrins, localizing urokinase to sites of cell attachment. Binding of uPAR to the beta-propeller of alpha3beta1 empowers vitronectin adhesion by this integrin. How uPAR modifies other beta1 integrins remains unknown. Using recombinant proteins, we found uPAR directly binds alpha5beta1 and rather than blocking, renders fibronectin (Fn) binding by alpha5beta1 Arg-Gly-Asp (RGD) resistant. This resulted from RGD-independent binding of alpha5beta1-uPAR to Fn type III repeats 12-15 in addition to type III repeats 9-11 bound by alpha5beta1. Suppression of endogenous uPAR by small interfering RNA in tumor cells promoted weaker, RGD-sensitive Fn adhesion and altered overall alpha5beta1 conformation. A beta1 peptide (res 224NLDSPEGGF232) that models near the known alpha-chain uPAR-binding region, or a beta1-chain Ser227Ala point mutation, abrogated effects of uPAR on alpha5beta1. Direct binding and regulation of alpha5beta1 by uPAR implies a modified "bent" integrin conformation can function in an alternative activation state with this and possibly other cis-acting membrane ligands.


Subject(s)
Cell Adhesion/physiology , Integrin alpha5beta1/metabolism , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cell Adhesion/drug effects , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Extracellular Matrix/metabolism , Fibronectins/metabolism , Gene Expression , Humans , Integrin alpha3beta1/genetics , Integrin alpha3beta1/metabolism , Integrin alpha5beta1/chemistry , Integrin alpha5beta1/genetics , Mice , Models, Molecular , Molecular Sequence Data , Mutation , Oligopeptides/pharmacology , Peptides/pharmacology , Plasminogen Activator Inhibitor 1/pharmacology , Protein Binding/drug effects , Protein Conformation , RNA, Small Interfering/genetics , Receptors, Cell Surface/genetics , Receptors, Immunologic/metabolism , Receptors, Peptide/metabolism , Receptors, Urokinase Plasminogen Activator , Recombinant Proteins/pharmacology , Sequence Alignment , Urokinase-Type Plasminogen Activator/pharmacology
13.
J Biol Chem ; 277(50): 48427-33, 2002 Dec 13.
Article in English | MEDLINE | ID: mdl-12376552

ABSTRACT

Growth hormone (GH) secretion is regulated by indirect negative feedback mechanisms. To address whether GH has direct actions on pituitary cells, lipid signaling in GH(4)ZR(7) somatomammotroph cells was examined. GH (EC(50) = 5 nm) stimulated diacylglycerol (DAG) and ceramide formation in parallel by over 10-fold within 15 min and persisting for >3 h. GH-induced DAG/ceramide formation was blocked by pertussis toxin (PTX) implicating G(i)/G(o) proteins and was potentiated 1.5-fold by activation of G(i)/G(o)-coupled dopamine-D2S receptors, which had no effect alone. Following PTX pretreatment, only PTX-resistant Galpha(i)3, not Galpha(o) or Galpha(i)2, rescued GH-induced DAG/ceramide signaling. GH-induced DAG/ceramide formation was also blocked in cells expressing Gbetagamma blocker GRK-ct. In GH(4)ZR(7) cells, GH induced phosphorylation of JAK2 and STAT5, which was blocked by PTX and mimicked by ceramide analogue C2-ceramide or sphingomyelinase treatment to increase endogenous ceramide. We conclude that in GH(4) pituitary cells, GH induces formation of DAG/ceramide via a novel Galpha(i)3/Gbetagamma-dependent pathway. This novel pathway suggests a mechanism for autocrine feedback regulation by GH of pituitary function.


Subject(s)
Ceramides/biosynthesis , Diglycerides/biosynthesis , GTP-Binding Proteins/metabolism , Growth Hormone/drug effects , Pituitary Gland/drug effects , Receptors, Dopamine D2/metabolism , Apomorphine/pharmacology , Cells, Cultured , Pertussis Toxin/pharmacology , Pituitary Gland/metabolism
14.
Cell Signal ; 14(5): 407-18, 2002 May.
Article in English | MEDLINE | ID: mdl-11882385

ABSTRACT

This review focuses on the coupling specificity of the Galpha and Gbetagamma subunits of pertussis toxin (PTX)-sensitive G(i/o) proteins that mediate diverse signaling pathways, including regulation of ion channels and other effectors. Several lines of evidence indicate that specific combinations of G protein alpha, beta and gamma subunits are required for different receptors or receptor-effector networks, and that a higher degree of specificity for Galpha and Gbetagamma is observed in intact systems than reported in vitro. The structural determinants of receptor-G protein specificity remain incompletely understood, and involve receptor-G protein interaction domains, and perhaps other scaffolding processes. By identifying G protein specificity for individual receptor signaling pathways, ligands targeted to disrupt individual pathways of a given receptor could be developed.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go/physiology , GTP-Binding Protein beta Subunits , GTP-Binding Protein gamma Subunits , Heterotrimeric GTP-Binding Proteins/physiology , Signal Transduction , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Animals , Calcium Channels/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , Heterotrimeric GTP-Binding Proteins/chemistry , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Macromolecular Substances , Models, Biological , Potassium Channels/metabolism , Receptors, Cell Surface/metabolism , Sequence Alignment
15.
J Biol Chem ; 277(9): 7348-55, 2002 Mar 01.
Article in English | MEDLINE | ID: mdl-11707461

ABSTRACT

Gbetagamma subunits interact directly and activate G protein-gated Inwardly Rectifying K(+) (GIRK) channels. Little is known about the identity of functionally important interactions between Gbetagamma and GIRK channels. We tested the effects of all mammalian Gbeta subunits on channel activity and showed that whereas Gbeta1-4 subunits activate heteromeric GIRK channels independently of receptor activation, Gbeta5 does not. Gbeta1 and Gbeta5 both bind the N and C termini of the GIRK1 and GIRK4 channel subunits. Chimeric analysis between the Gbeta1 and Gbeta5 proteins revealed a 90-amino acid stretch that spans blades two and three of the seven-propeller structure and is required for channel activation. Within this region, eight non-conserved amino acids were critical for the activity of Gbeta1, as mutation of each residue to its counterpart in Gbeta5 significantly reduced the ability of Gbeta1 to stimulate channel activity. In particular, mutation of residues Ser-67 and Thr-128 to the corresponding Gbeta5 residues completely abolished Gbeta1 stimulation of GIRK channel activity. Mapping these functionally important residues on the three-dimensional structure of Gbeta1 shows that Ser-67, Ser-98, and Thr-128 are the only surface accessible residues. Galpha(i)1 interacts with Ser-98 but not with Ser-67 and Thr-128 in the heterotrimeric Galphabetagamma structure. Further characterization of the three mutant proteins showed that they fold properly and interact with Ggamma2. Of the three identified functionally important residues, the Ser-67 and Thr-128 Gbeta mutants significantly inhibited basal currents of a channel point mutant that displays Gbetagamma-mediated basal but not agonist-induced currents. Our findings indicate that the presence of Gbeta residues that do not interact with Galpha are involved in Gbetagamma interactions in the absence of agonist stimulation.


Subject(s)
GTP-Binding Protein beta Subunits , Heterotrimeric GTP-Binding Proteins/chemistry , Potassium Channels, Inwardly Rectifying , Potassium Channels/metabolism , Amino Acid Sequence , Amino Acids/chemistry , Animals , DNA, Complementary/metabolism , Electrophysiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels , Glutathione Transferase/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Immunoblotting , Insecta , Kinetics , Models, Molecular , Molecular Sequence Data , Mutation , Oocytes/metabolism , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Serine/chemistry , Signal Transduction , Threonine/chemistry , Time Factors , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL