Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
J Biol Chem ; 299(7): 104887, 2023 07.
Article in English | MEDLINE | ID: mdl-37271338

ABSTRACT

The neuroepithelial cell transforming gene 1 (Net1) is a guanine nucleotide exchange factor for the small GTPase RhoA that promotes cancer cell motility and metastasis. Two isoforms of Net1 exist, Net1 and Net1A, both of which are sequestered in the nucleus in quiescent cells to prevent aberrant RhoA activation. Many cell motility stimuli drive cytosolic relocalization of Net1A, but mechanisms controlling this event are not fully understood. Here, we demonstrate that epithelial growth factor stimulates protein kinase Src- and Abl1-dependent phosphorylation of Net1A to promote its cytosolic localization. We show that Abl1 efficiently phosphorylates Net1A on Y373, and that phenylalanine substitution of Y373 prevents Net1A cytosolic localization. Furthermore, we found that Abl1-driven cytosolic localization of Net1A does not require S52, which is a phosphorylation site of a different kinase, c-Jun N-terminal kinase, that inhibits nuclear import of Net1A. However, we did find that MKK7-stimulated cytosolic localization of Net1A does require Y373. We also demonstrate that aspartate substitution at Y373 is sufficient to promote Net1A cytosolic accumulation, and expression of Net1A Y373D potentiates epithelial growth factor-stimulated RhoA activation, downstream myosin light chain 2 phosphorylation, and F-actin accumulation. Moreover, we show that expression of Net1A Y373D in breast cancer cells also significantly increases cell motility and Matrigel invasion. Finally, we show that Net1A is required for Abl1-stimulated cell motility, which is rescued by expression of Net1A Y373D, but not Net1A Y373F. Taken together, this work demonstrates a novel mechanism controlling Net1A subcellular localization to regulate RhoA-dependent cell motility and invasion.


Subject(s)
Guanine Nucleotide Exchange Factors , Proto-Oncogene Proteins c-abl , rhoA GTP-Binding Protein , Cell Movement , Cytosol/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Phosphorylation , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism , Proto-Oncogene Proteins c-abl/metabolism
2.
J Biol Chem ; 299(5): 104645, 2023 05.
Article in English | MEDLINE | ID: mdl-36965619

ABSTRACT

The Somatostatin receptor 2 (Sstr2) is a heterotrimeric G protein-coupled receptor that is highly expressed in neuroendocrine tumors and is a common pharmacological target for intervention. Unfortunately, not all neuroendocrine tumors express Sstr2, and Sstr2 expression can be downregulated with prolonged agonist use. Sstr2 is rapidly internalized following agonist stimulation and, in the short term, is quantitatively recycled back to the plasma membrane. However, mechanisms controlling steady state expression of Sstr2 in the absence of agonist are less well described. Here, we show that Sstr2 interacts with the Wnt pathway protein Dvl1 in a ligand-independent manner to target Sstr2 for lysosomal degradation. Interaction of Sstr2 with Dvl1 does not affect receptor internalization, recycling, or signaling to adenylyl cyclase but does suppress agonist-stimulated ERK1/2 activation. Importantly, Dvl1-dependent degradation of Sstr2 can be stimulated by overexpression of Wnts and treatment of cells with Wnt pathway inhibitors can boost Sstr2 expression in neuroendocrine tumor cells. Taken together, this study identifies for the first time a mechanism that targets Sstr2 for lysosomal degradation that is independent of Sstr2 agonist and can be potentiated by Wnt ligand. Intervention in this signaling mechanism has the potential to elevate Sstr2 expression in neuroendocrine tumors and enhance Sstr2-directed therapies.


Subject(s)
Dishevelled Proteins , Lysosomes , Receptors, Somatostatin , Humans , Dishevelled Proteins/genetics , Dishevelled Proteins/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , Lysosomes/metabolism , Neuroendocrine Tumors/physiopathology , Protein Binding , Protein Transport , Receptors, Somatostatin/genetics , Receptors, Somatostatin/metabolism
3.
ACS Bio Med Chem Au ; 2(6): 617-626, 2022 Dec 21.
Article in English | MEDLINE | ID: mdl-37101428

ABSTRACT

We describe a small molecule ligand ACA-14 (2-hydroxy-5-{[(2-phenylcyclopropyl) carbonyl] amino} benzoic acid) as an initial lead for the development of direct inhibitors of KRAS, a notoriously difficult anticancer drug target. We show that the compound binds to KRAS near the switch regions with affinities in the low micromolar range and exerts different effects on KRAS interactions with binding partners. Specifically, ACA-14 impedes the interaction of KRAS with its effector Raf and reduces both intrinsic and SOS-mediated nucleotide exchange rates. Likely as a result of these effects, ACA-14 inhibits signal transduction through the MAPK pathway in cells expressing mutant KRAS and inhibits the growth of pancreatic and colon cancer cells harboring mutant KRAS. We thus propose compound ACA-14 as a useful initial lead for the development of broad-acting inhibitors that target multiple KRAS mutants and simultaneously deplete the fraction of GTP-loaded KRAS while abrogating the effector-binding ability of the already GTP-loaded fraction.

5.
Eur J Med Chem ; 217: 113381, 2021 May 05.
Article in English | MEDLINE | ID: mdl-33756124

ABSTRACT

KRAS plays an essential role in regulating cell proliferation, differentiation, migration and survival. Mutated KRAS is a major driver of malignant transformation in multiple human cancers. We showed previously that fendiline (6) is an effective inhibitor of KRAS plasma membrane (PM) localization and function. In this study, we designed, synthesized and evaluated a series of new fendiline analogs to optimize its drug properties. Systemic structure-activity relationship studies by scaffold repurposing led to the discovery of several more active KRAS PM localization inhibitors such as compounds 12f (NY0244), 12h (NY0331) and 22 (NY0335) which exhibit nanomolar potencies. These compounds inhibited oncogenic KRAS-driven cancer cell proliferation at single-digit micromolar concentrations in vitro. In vivo studies in a xenograft model of pancreatic cancer revealed that 12h and 22 suppressed oncogenic KRAS-expressing MiaPaCa-2 tumor growth at a low dose range of 1-5 mg/kg with no vasodilatory effects, indicating their potential as chemical probes and anticancer therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Membrane/drug effects , Enzyme Inhibitors/pharmacology , Fendiline/pharmacology , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Dogs , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Female , Fendiline/analogs & derivatives , Fendiline/chemistry , Humans , Mice , Mice, Nude , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Structure-Activity Relationship
6.
Cell Signal ; 80: 109926, 2021 04.
Article in English | MEDLINE | ID: mdl-33465404

ABSTRACT

The Neuroepithelial transforming gene 1 (Net1) is a RhoA subfamily guanine nucleotide exchange factor that is overexpressed in a number of cancers and contributes to cancer cell motility and proliferation. Net1 also plays a Rho GTPase independent role in mitotic progression, where it promotes centrosomal activation of Aurora A and Pak2, and aids in chromosome alignment during prometaphase. To understand regulatory mechanisms controlling the mitotic function of Net1, we examined whether it was phosphorylated by the mitotic kinase Cdk1. We observed that Cdk1 phosphorylated Net1 on multiple sites in its N-terminal regulatory domain and C-terminus in vitro. By raising phospho-specific antibodies to two of these sites, we also demonstrated that both endogenous and transfected Net1 were phosphorylated by Cdk1 in cells. Substitution of the major Cdk1 phosphorylation sites with aliphatic or acidic residues inhibited the interaction of Net1 with RhoA, and treatment of metaphase cells with a Cdk1 inhibitor increased Net1 activity. Cdk1 inhibition also increased Net1 localization to the plasma membrane and stimulated cortical F-actin accumulation. Moreover, Net1 overexpression caused spindle polarity defects that were reduced in frequency by acidic substitution of the major Cdk1 phosphorylation sites. These data indicate that Cdk1 phosphorylates Net1 during mitosis and suggest that this negatively regulates its ability to signal to RhoA and alter actin cytoskeletal organization.


Subject(s)
CDC2 Protein Kinase/metabolism , Mitosis , Oncogene Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Actin Cytoskeleton , Actins/metabolism , CDC2 Protein Kinase/antagonists & inhibitors , CDC2 Protein Kinase/genetics , Cell Membrane/metabolism , HeLa Cells , Humans , Mutagenesis, Site-Directed , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/genetics , Phosphorylation , Protein Stability , RNA Interference , RNA, Small Interfering/metabolism , Spindle Apparatus/physiology , rhoA GTP-Binding Protein/genetics
7.
Endocrinology ; 162(2)2021 02 01.
Article in English | MEDLINE | ID: mdl-33313679

ABSTRACT

The somatostatin receptor 2A (SST2) is a G-protein-coupled receptor (GPCR) that is expressed in neuroendocrine tissues within the gastrointestinal tract and brain, and is commonly overexpressed in many neuroendocrine tumors. Moreover, SST2 agonists are used clinically as the primary pharmacological treatment to suppress excess hormone secretion in a variety of neuroendocrine tumors. Despite its wide clinical use, mechanisms controlling the trafficking and signaling of SST2 are not fully understood. SST2 contains a C-terminal post-synaptic density 95, Drosophila discs large, zona-occludens 1 (PDZ) domain-binding motif that has been shown to interact with 3 different PDZ domain-containing proteins. However, the consequences of these interactions are not well understood, nor is it known whether additional PDZ domain proteins interact with SST2. Through unbiased screening we have identified 10 additional PDZ domain proteins that interact with SST2. We chose one of these, SYNJ2BP, for further study. We observed that SYNJ2BP interacted with SST2 in an agonist-dependent manner, and that this required the PDZ binding site of SST2. Importantly, overexpression of SYNJ2BP enhanced ligand-stimulated receptor internalization. Mechanistically, SYNJ2BP interacted with G-protein-coupled receptor kinase 2 (GRK2) and promoted GRK-dependent phosphorylation of the receptor after somatostatin stimulation. Interaction with GRK2 required the C-terminus of SYNJ2BP. Binding to SYNJ2BP did not affect the ability of SST2 to suppress 3',5'-cyclic adenosine 5'-monophosphate production, but was required for optimal agonist-stimulated extracellularly regulated kinase 1/2 activation. These data indicated that SYNJ2BP is an SST2-interacting protein that modulates agonist-stimulated receptor regulation and downstream signaling.


Subject(s)
G-Protein-Coupled Receptor Kinase 2/metabolism , MAP Kinase Signaling System , Membrane Proteins/metabolism , PDZ Domains , Receptors, Somatostatin/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Phosphorylation
8.
Cancer Res ; 81(4): 885-897, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33355181

ABSTRACT

Cancer-associated cachexia, characterized by muscle wasting, is a lethal metabolic syndrome without defined etiology or established treatment. We previously found that p300 mediates cancer-induced muscle wasting by activating C/EBPß, which then upregulates key catabolic genes. However, the signaling mechanism that activates p300 in response to cancer is unknown. Here, we show that upon cancer-induced activation of Toll-like receptor 4 in skeletal muscle, p38ß MAPK phosphorylates Ser-12 on p300 to stimulate C/EBPß acetylation, which is necessary and sufficient to cause muscle wasting. Thus, p38ß MAPK is a central mediator and therapeutic target of cancer-induced muscle wasting. In addition, nilotinib, an FDA-approved kinase inhibitor that preferentially binds p38ß MAPK, inhibited p300 activation 20-fold more potently than the p38α/ß MAPK inhibitor, SB202190, and abrogated cancer cell-induced muscle protein loss in C2C12 myotubes without suppressing p38α MAPK-dependent myogenesis. Systemic administration of nilotinib at a low dose (0.5 mg/kg/day, i.p.) in tumor-bearing mice not only alleviated muscle wasting, but also prolonged survival. Therefore, nilotinib appears to be a promising treatment for human cancer cachexia due to its selective inhibition of p38ß MAPK. SIGNIFICANCE: These findings demonstrate that prevention of p38ß MAPK-mediated activation of p300 by the FDA-approved kinase inhibitor, nilotinib, ameliorates cancer cachexia, representing a potential therapeutic strategy against this syndrome.


Subject(s)
Cachexia/etiology , Mitogen-Activated Protein Kinase 11/metabolism , Muscular Atrophy/etiology , Neoplasms/complications , Animals , Cachexia/genetics , Cachexia/metabolism , Cachexia/pathology , Carcinoma, Lewis Lung/complications , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/pathology , Cells, Cultured , Humans , Imidazoles/pharmacology , Lung Neoplasms/complications , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 11/antagonists & inhibitors , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation/drug effects , Pyridines/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology
9.
Small GTPases ; 11(4): 293-300, 2020 07.
Article in English | MEDLINE | ID: mdl-29173011

ABSTRACT

Macrophages are innate immune cells that constantly patrol an organism to fulfill protective and homeostatic roles. Previous studies have shown that Rho GTPase activity is required for macrophage mobility, yet the roles of upstream regulatory proteins controlling Rho GTPase function in these cells are not well defined. Previously we have shown that the RhoA GEF Net1 is required for human breast cancer cell motility and extracellular matrix invasion. To assess the role of Net1 in macrophage motility, we isolated bone marrow macrophage (BMM) precursors from wild type and Net1 knockout mice. Loss of Net1 did not affect the ability of BMM precursors to differentiate into mature macrophages in vitro, as measured by CD68 and F4/80 staining. However, Net1 deletion significantly reduced RhoA activation, F-actin accumulation, adhesion, and motility in these cells. Nevertheless, similar to RhoA/RhoB double knockout macrophages, Net1 deletion did not impair macrophage recruitment to the peritoneum in a mouse model of sterile inflammation. These data demonstrate that Net1 is an important regulator of RhoA signaling and motility in mouse macrophages in vitro, but that its function may be dispensable for macrophage recruitment to inflammatory sites in vivo.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Macrophages/metabolism , Oncogene Proteins/genetics , Rho Guanine Nucleotide Exchange Factors/genetics , Animals , Cell Differentiation , Cells, Cultured , Gene Deletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Oncogene Proteins/deficiency , Oncogene Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism
10.
Small GTPases ; 11(6): 385-391, 2020 11.
Article in English | MEDLINE | ID: mdl-30332929

ABSTRACT

Jnks are mitogen activated protein kinases that are best known for regulating transcription and apoptotic signaling. However, they also play important roles in controlling cell motility and invasion by phosphorylating many actin and microtubule regulatory proteins. These mechanisms have important implications for normal cell motility as well as cancer metastasis. Jnks are activated by growth factors and cytokines that stimulate cell motility, and this often requires upstream activation of Rho GTPases. Our recent work indicates that Jnks may also regulate Rho GTPase activation. Specifically, we found that Jnk-dependent phosphorylation of the RhoA guanine nucleotide exchange factor (RhoGEF) Net1A promotes its cytosolic accumulation to drive RhoA activation and actin cytoskeletal reorganization. Net1A is unusual among RhoGEFs in that it is sequestered in the nucleus to prevent aberrant RhoA activation. Importantly, Jnk-stimulated cytosolic localization of Net1A is sufficient to stimulate cell motility and extracellular matrix invasion in non-invasive breast cancer cells. Since Net1A expression is critical for cancer cell motility and invasion in vitro, and breast cancer metastasis in vivo, these data uncover a previously unappreciated regulatory mechanism that may contribute to metastasis in multiple types of cancer.


Subject(s)
JNK Mitogen-Activated Protein Kinases/metabolism , Oncogene Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Cell Movement , Humans
11.
Endocrinology ; 160(5): 1031-1043, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30822353

ABSTRACT

The Gi-coupled somatostatin receptor 2 (SST2) is a G protein-coupled receptor (GPCR) that mediates many of somatostatin's neuroendocrine actions. Upon stimulation, SST2 is rapidly internalized and transported to early endosomes before being recycled to the plasma membrane. However, little is known about the intracellular itinerary of SST2 after it moves to the early endosomal compartment or the cytoplasmic proteins that regulate its trafficking. As postsynaptic density protein/discs large 1/zonula occludens-1 (PDZ) domain interactions often regulate the trafficking and signaling potential of GPCRs, we examined the role of the SST2 PDZ ligand and additional C-terminal residues in controlling its intracellular trafficking. We determined that SST2 can recycle to the plasma membrane via multiple pathways, including a LAMP1/Rab7-positive late endosome to the trans-Golgi network (TGN) pathway. Trafficking from the late endosome to the TGN is often regulated by the retromer complex of endosomal coat proteins, and disrupting the retromer components sorting nexins 1/2 inhibits the budding of SST2 from late endosomes. Moreover, trafficking through the late endosomal/TGN pathway is dependent on an intact PDZ ligand and C-terminal tail, as truncating either the 3 or 10 C-terminal amino acids of SST2 alters the pathway through which it recycles to the plasma membrane. Moreover, addition of these amino acids to a heterologous receptor is sufficient to redirect it from a degradation pathway to a recycling itinerary. Our results demonstrate that endosomal trafficking of SST2 is dependent on numerous regulatory mechanisms controlled by its C terminus and the retromer machinery.


Subject(s)
Cell Membrane/metabolism , Endosomes/metabolism , Membrane Proteins/metabolism , Receptors, Somatostatin/metabolism , trans-Golgi Network/metabolism , Amino Acid Motifs , Amino Acid Sequence , Base Sequence , HEK293 Cells , Humans , Multiprotein Complexes/metabolism , Nucleotide Motifs , PDZ Domains , Protein Transport , Receptors, Somatostatin/chemistry , Receptors, Somatostatin/genetics , Signal Transduction
12.
J Pain ; 20(5): 528-539, 2019 05.
Article in English | MEDLINE | ID: mdl-30471428

ABSTRACT

Paclitaxel-induced peripheral neuropathy (PIPN) and associated neuropathic pain are the most common and serious adverse effects experienced by cancer patients receiving paclitaxel treatment. These effects adversely impact daily activities and consequently the quality of life, sometimes forcing the suspension of treatment and negatively influencing survival. Patients are usually at high risk of developing PIPN if paclitaxel induces acute pain, which strongly suggests that an acute increase in the excitability of nociceptors underlies the chronic alterations of PIPN. KCNQ/Kv7 channels are widely expressed in the primary sensory neurons to modulate their excitability. In the present study, we show that targeting KCNQ/Kv7 channels at an early stage is an effective strategy to attenuate the development of PIPN. We found that paclitaxel did not decrease the expression level of KCNQ/Kv7 channels in the primary sensory neurons as detected by quantitative reverse-transcription polymerase chain reaction (qRT-PCR) and Western blotting. However, retigabine, which is a specific KCNQ/Kv7 channel opener, attenuated significantly the development of PIPN, as shown by both morphologic and behavioral evidence. We also observed that retigabine had no obvious effect on the chemosensitivity of breast cancer cells to paclitaxel. Although retigabine has been approved by the FDA as an anticonvulsant, our study suggests that this drug can be repurposed to attenuate the development of PIPN. PERSPECTIVE: Paclitaxel-induced peripheral neuropathy and associated neuropathic pain are severe and resistant to intervention. The results of our study demonstrated that retigabine (a clinically available medicine) can be used to attenuate the development of paclitaxel-induced peripheral neuropathy.


Subject(s)
Carbamates/pharmacology , KCNQ Potassium Channels/agonists , Neuralgia/prevention & control , Neuroprotective Agents/pharmacology , Paclitaxel/adverse effects , Peripheral Nervous System Diseases/prevention & control , Phenylenediamines/pharmacology , Animals , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Drug Repositioning , Humans , KCNQ Potassium Channels/metabolism , Male , Neuralgia/chemically induced , Neuralgia/metabolism , Neurons/drug effects , Neurons/metabolism , Paclitaxel/pharmacology , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/metabolism , Random Allocation , Rats, Sprague-Dawley
13.
J Endocr Soc ; 2(7): 779-793, 2018 Jul 01.
Article in English | MEDLINE | ID: mdl-30151433

ABSTRACT

Acromegaly is a neuroendocrine disorder caused by excess secretion of GH by somatotroph tumor cells. It is often treated with somatostatin receptor (SSTR) 2 agonists, which suppress GH secretion. SOM230 is a somatostatin analogue that targets multiple SSTRs and was recently approved for patients with treatment-resistant acromegaly. Previous reports indicate that SOM230 may function as a biased agonist, suggesting that its ability to selectively activate SSTR-dependent signaling events may contribute to its therapeutic efficacy. To better understand how SOM230 modulates Sstr2A function, which is the most commonly expressed SSTR in somatotrophs, we used real-time assays to study SOM230-dependent signaling in rat pituitary tumor cells. We observed that SOM230 suppressed cAMP production in a Gαi-dependent manner, similar to conventional Sstr2A agonists. However, it did not cause receptor internalization as would be expected for an Sstr2A agonist. Surprisingly, SOM230 did not cause membrane hyperpolarization, which is an important mechanism by which Sstr2a activation suppresses intracellular calcium (Ca2+) accumulation and GH secretion. In fact, SOM230 inhibited the ability of conventional somatostatin analogues to control membrane potential. However, SOM230 still inhibited intracellular Ca2+ accumulation in a novel, Gßγ-dependent manner. These studies show that SOM230 exhibits strong agonist bias in regulating signaling pathways downstream of Sstr2A that control GH secretion.

14.
Breast Cancer Res ; 20(1): 41, 2018 05 16.
Article in English | MEDLINE | ID: mdl-29769144

ABSTRACT

BACKGROUND: The RhoA activating protein Net1 contributes to breast cancer cell proliferation, motility, and invasion in vitro, yet little is known about its roles in mammary gland tumorigenesis and metastasis. METHODS: Net1 knockout (KO) mice were bred to mice with mammary gland specific expression of the polyoma middle T antigen (PyMT) oncogene. Mammary gland tumorigenesis and lung metastasis were monitored. Individual tumors were assessed for proliferation, apoptosis, angiogenesis, RhoA activation, and activation of PyMT-dependent signaling pathways. Primary tumor cells from wild-type and Net1 KO mice were transplanted into the mammary glands of wild-type, nontumor-bearing mice, and tumor growth and metastasis were assessed. Gene expression in wild-type and Net1 KO tumors was analyzed by gene ontology enrichment and for relative activation of gene expression signatures indicative of signaling pathways important for breast cancer initiation and progression. A gene expression signature indicative of Net1 function was identified. Human breast cancer gene expression profiles were screened for the presence of a Net1 gene expression signature. RESULTS: We show that Net1 makes fundamental contributions to mammary gland tumorigenesis and metastasis. Net1 deletion delays tumorigenesis and strongly suppresses metastasis in PyMT-expressing mice. Moreover, we observe that loss of Net1 reduces cancer cell proliferation, inhibits tumor angiogenesis, and promotes tumor cell apoptosis. Net1 is required for maximal RhoA activation within tumors and for primary tumor cell motility. Furthermore, the ability of PyMT to initiate oncogenic signaling to ERK1/2 and PI3K/Akt1 is inhibited by Net1 deletion. Primary tumor cell transplantation indicates that the reduction in tumor angiogenesis and lung metastasis observed upon Net1 deletion are tumor cell autonomous effects. Using a gene expression signature indicative of Net1 activity, we show that Net1 signaling is activated in 10% of human breast cancers, and that this correlates with elevated proliferation and PI3K pathway activity. We also demonstrate that human breast cancer patients with a high Net1 gene expression signature experience shorter distant metastasis-free survival. CONCLUSIONS: These data indicate that Net1 is required for tumor progression in the PyMT mouse model and suggest that Net1 may contribute to breast cancer progression in humans.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Breast Neoplasms/genetics , Lung Neoplasms/genetics , Mammary Neoplasms, Animal/genetics , Oncogene Proteins/genetics , Animals , Breast Neoplasms/pathology , Carcinogenesis/genetics , Cell Movement , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mammary Glands, Human/pathology , Mammary Neoplasms, Animal/pathology , Mice , Mice, Knockout , Neoplasm Metastasis , rhoA GTP-Binding Protein/genetics
15.
J Cell Sci ; 131(3)2018 02 01.
Article in English | MEDLINE | ID: mdl-29361525

ABSTRACT

The neuroepithelial cell transforming gene 1A (Net1A, an isoform of Net1) is a RhoA subfamily guanine nucleotide exchange factor (GEF) that localizes to the nucleus in the absence of stimulation, preventing it from activating RhoA. Once relocalized in the cytosol, Net1A stimulates cell motility and extracellular matrix invasion. In the present work, we investigated mechanisms responsible for the cytosolic relocalization of Net1A. We demonstrate that inhibition of MAPK pathways blocks Net1A relocalization, with cells being most sensitive to JNK pathway inhibition. Moreover, activation of the JNK or p38 MAPK family pathway is sufficient to elicit Net1A cytosolic localization. Net1A relocalization stimulated by EGF or JNK activation requires nuclear export mediated by CRM1. JNK1 (also known as MAPK8) phosphorylates Net1A on serine 52, and alanine substitution at this site prevents Net1A relocalization caused by EGF or JNK activation. Glutamic acid substitution at this site is sufficient for Net1A relocalization and results in elevated RhoA signaling to stimulate myosin light chain 2 (MLC2, also known as MYL2) phosphorylation and F-actin accumulation. Net1A S52E expression stimulates cell motility, enables Matrigel invasion and promotes invadopodia formation. These data highlight a novel mechanism for controlling the subcellular localization of Net1A to regulate RhoA activation, cell motility, and invasion.


Subject(s)
Cell Movement , Karyopherins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Oncogene Proteins/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Stress, Physiological , Cell Movement/drug effects , Cytosol/drug effects , Cytosol/metabolism , Epidermal Growth Factor/pharmacology , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Glutamic Acid/metabolism , Humans , MCF-7 Cells , Mutation/genetics , Phosphorylation/drug effects , Phosphoserine/metabolism , Podosomes/drug effects , Podosomes/metabolism , Protein Transport/drug effects , Signal Transduction , Stress, Physiological/drug effects , Tumor Necrosis Factor-alpha/pharmacology , rhoA GTP-Binding Protein/metabolism , Exportin 1 Protein
16.
Mol Cell Biol ; 38(3)2018 02 01.
Article in English | MEDLINE | ID: mdl-29158292

ABSTRACT

K-Ras must localize to the plasma membrane (PM) for biological activity. We show here that multiple acid sphingomyelinase (ASM) inhibitors, including tricyclic antidepressants, mislocalized phosphatidylserine (PtdSer) and K-RasG12V from the PM, resulting in abrogation of K-RasG12V signaling and potent, selective growth inhibition of mutant K-Ras-transformed cancer cells. Concordantly, in nude mice, the ASM inhibitor fendiline decreased the rate of growth of oncogenic K-Ras-expressing MiaPaCa-2 tumors but had no effect on the growth of the wild-type K-Ras-expressing BxPC-3 tumors. ASM inhibitors also inhibited activated LET-60 (a K-Ras ortholog) signaling in Caenorhabditis elegans, as evidenced by suppression of the induced multivulva phenotype. Using RNA interference against C. elegans genes encoding other enzymes in the sphingomyelin (SM) biosynthetic pathway, we identified 14 enzymes whose knockdown strongly or moderately suppressed the LET-60 multivulva phenotype. In mammalian cells, pharmacological agents that target these enzymes all depleted PtdSer from the PM and caused K-RasG12V mislocalization. These effects correlated with changes in SM levels or subcellular distribution. Selected compounds, including sphingosine kinase inhibitors, potently inhibited the proliferation of oncogenic K-Ras-expressing pancreatic cancer cells. In conclusion, these results show that normal SM metabolism is critical for K-Ras function, which may present therapeutic options for the treatment of K-Ras-driven cancers.


Subject(s)
Sphingolipids/metabolism , Sphingomyelin Phosphodiesterase/metabolism , Animals , Caenorhabditis elegans , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation , Humans , Mice , Mice, Nude , Signal Transduction , Sphingomyelins/genetics , Sphingomyelins/metabolism , ras Proteins/metabolism
17.
J Neurotrauma ; 34(6): 1260-1270, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28073317

ABSTRACT

A majority of people who have sustained spinal cord injury (SCI) experience chronic pain after injury, and this pain is highly resistant to available treatments. Contusive SCI in rats at T10 results in hyperexcitability of primary sensory neurons, which contributes to chronic pain. KCNQ channels are widely expressed in nociceptive dorsal root ganglion (DRG) neurons, are important for controlling their excitability, and their activation has proven effective in reducing pain in peripheral nerve injury and inflammation models. The possibility that activators of KCNQ channels could be useful for treating SCI-induced chronic pain is strongly supported by the following findings. First, SCI, unlike peripheral nerve injury, failed to decrease the functional or biochemical expression of KCNQ channels in DRG as revealed by electrophysiology, real-time quantitative polymerase chain reaction, and Western blot; therefore, these channels remain available for pharmacological targeting of SCI pain. Second, treatment with retigabine, a specific KCNQ channel opener, profoundly decreased spontaneous activity in primary sensory neurons of SCI animals both in vitro and in vivo without changing the peripheral mechanical threshold. Third, retigabine reversed SCI-induced reflex hypersensitivity, adding to our previous demonstration that retigabine supports the conditioning of place preference after SCI (an operant measure of spontaneous pain). In contrast to SCI animals, naïve animals showed no effects of retigabine on reflex sensitivity or conditioned place preference by pairing with retigabine, indicating that a dose that blocks chronic pain-related behavior has no effect on normal pain sensitivity or motivational state. These results encourage the further exploration of U.S. Food and Drug Administration-approved KCNQ activators for treating SCI pain, as well as efforts to develop a new generation of KCNQ activators that lack central side effects.


Subject(s)
Behavior, Animal/drug effects , Carbamates/pharmacology , Chronic Pain/metabolism , Ganglia, Spinal/metabolism , KCNQ Potassium Channels/metabolism , Membrane Transport Modulators/pharmacology , Phenylenediamines/pharmacology , Spinal Cord Injuries/metabolism , Animals , Carbamates/administration & dosage , Chronic Pain/drug therapy , Disease Models, Animal , Ganglia, Spinal/drug effects , KCNQ Potassium Channels/drug effects , Male , Membrane Transport Modulators/administration & dosage , Phenylenediamines/administration & dosage , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/drug therapy
18.
Small GTPases ; 7(2): 76-81, 2016 04 02.
Article in English | MEDLINE | ID: mdl-27070728

ABSTRACT

Rho GTPases regulate cell motility in a large part through control of actin cytoskeletal organization. The activation state of Rho proteins is regulated by a wide variety of guanine nucleotide exchange factors (GEFs) and GTPase activating proteins that are differentially expressed among cell types and disease states. The RhoA specific GEF neuroepithelial transforming gene 1 (Net1) is highly expressed in many cancer cells and stimulates cell motility, invasion and cell spreading in response to a variety of ligands. A key feature of Net1 proteins is that they are sequestered in the nucleus in the absence of a motility stimulus. We have recently found that accumulation of the Net1A isoform outside the nucleus, which is the primary Net1 isoform controlling cell motility, is regulated by its acetylation status. Here, we describe acetylation as a novel mechanism of RhoGEF regulation in cell motility that can be targeted in cancer and metastasis.


Subject(s)
Cytoskeleton/metabolism , rhoA GTP-Binding Protein/metabolism , Acetylation , Cell Nucleus/metabolism , Enzyme Activation , Humans
19.
Mol Endocrinol ; 30(3): 278-89, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26677753

ABSTRACT

Ras homolog (Rho) family small GTPases are critical regulators of actin cytoskeletal organization, cell motility, proliferation, and survival. Surprisingly, the large majority of the studies underlying our knowledge of Rho protein function have been carried out in cultured cells, and it is only recently that researchers have begun to assess Rho GTPase regulation and function in vivo. The purpose of this review is to evaluate our current knowledge of Rho GTPase function in mouse mammary gland development, tumorigenesis and metastasis. Although our knowledge is still incomplete, these studies are already uncovering important themes as to the physiological roles of Rho GTPase signaling in normal mammary gland development and function. Essential contributions of Rho proteins to breast cancer initiation, tumor progression, and metastatic dissemination have also been identified.


Subject(s)
Carcinogenesis/metabolism , Carcinogenesis/pathology , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/pathology , rho GTP-Binding Proteins/metabolism , Animals , Disease Models, Animal , Female , Mice , Neoplasm Metastasis
20.
J Cell Sci ; 128(5): 913-22, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25588829

ABSTRACT

Net1 isoform A (Net1A) is a RhoA GEF that is required for cell motility and invasion in multiple cancers. Nuclear localization of Net1A negatively regulates its activity, and we have recently shown that Rac1 stimulates Net1A relocalization to the plasma membrane to promote RhoA activation and cytoskeletal reorganization. However, mechanisms controlling the subcellular localization of Net1A are not well understood. Here, we show that Net1A contains two nuclear localization signal (NLS) sequences within its N-terminus and that residues surrounding the second NLS sequence are acetylated. Treatment of cells with deacetylase inhibitors or expression of active Rac1 promotes Net1A acetylation. Deacetylase inhibition is sufficient for Net1A relocalization outside the nucleus, and replacement of the N-terminal acetylation sites with arginine residues prevents cytoplasmic accumulation of Net1A caused by deacetylase inhibition or EGF stimulation. By contrast, replacement of these sites with glutamine residues is sufficient for Net1A relocalization, RhoA activation and downstream signaling. Moreover, the N-terminal acetylation sites are required for rescue of F-actin accumulation and focal adhesion maturation in Net1 knockout MEFs. These data indicate that Net1A acetylation regulates its subcellular localization to impact on RhoA activity and actin cytoskeletal organization.


Subject(s)
Cell Membrane/metabolism , Cell Nucleus/metabolism , Cytoskeleton/metabolism , Oncogene Proteins/metabolism , Acetylation , Active Transport, Cell Nucleus/physiology , Animals , Cell Membrane/genetics , Cell Nucleus/genetics , Cytoskeleton/genetics , HeLa Cells , Humans , Mice , Mice, Knockout , Neuropeptides/genetics , Neuropeptides/metabolism , Nuclear Localization Signals/genetics , Nuclear Localization Signals/metabolism , Oncogene Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...