Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.280
Filter
1.
Annu Rev Cell Dev Biol ; 34: 29-58, 2018 10 06.
Article in English | MEDLINE | ID: mdl-30110558

ABSTRACT

Cell adhesion to macromolecules in the microenvironment is essential for the development and maintenance of tissues, and its dysregulation can lead to a range of disease states, including inflammation, fibrosis, and cancer. The biomechanical and biochemical mechanisms that mediate cell adhesion rely on signaling by a range of effector proteins, including kinases and associated scaffolding proteins. The intracellular trafficking of these must be tightly controlled in space and time to enable effective cell adhesion and microenvironmental sensing and to integrate cell adhesion with, and compartmentalize it from, other cellular processes, such as gene transcription, protein degradation, and cell division. Delivery of adhesion receptors and signaling proteins from the plasma membrane to unanticipated subcellular locales is revealing novel biological functions. Here, we review the expected and unexpected trafficking, and sites of activity, of adhesion and growth factor receptors and intracellular kinase partners as we begin to appreciate the complexity and diversity of their spatial regulation.


Subject(s)
Cell Adhesion/genetics , Platelet Glycoprotein GPIb-IX Complex/genetics , Protein Transport/genetics , Receptors, Growth Factor/genetics , Cell Membrane/genetics , Cell Nucleus/genetics , Endosomes/genetics , Humans , Phosphotransferases/genetics
2.
Mol Cell ; 80(1): 164-174.e4, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32877642

ABSTRACT

SARS-CoV-2 infections are rapidly spreading around the globe. The rapid development of therapies is of major importance. However, our lack of understanding of the molecular processes and host cell signaling events underlying SARS-CoV-2 infection hinders therapy development. We use a SARS-CoV-2 infection system in permissible human cells to study signaling changes by phosphoproteomics. We identify viral protein phosphorylation and define phosphorylation-driven host cell signaling changes upon infection. Growth factor receptor (GFR) signaling and downstream pathways are activated. Drug-protein network analyses revealed GFR signaling as key pathways targetable by approved drugs. The inhibition of GFR downstream signaling by five compounds prevents SARS-CoV-2 replication in cells, assessed by cytopathic effect, viral dsRNA production, and viral RNA release into the supernatant. This study describes host cell signaling events upon SARS-CoV-2 infection and reveals GFR signaling as a central pathway essential for SARS-CoV-2 replication. It provides novel strategies for COVID-19 treatment.


Subject(s)
Antiviral Agents/therapeutic use , Betacoronavirus/drug effects , Mitogen-Activated Protein Kinases/genetics , Phosphatidylinositol 3-Kinase/genetics , Receptors, Growth Factor/genetics , Viral Proteins/genetics , Adrenal Cortex Hormones/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Antibodies, Neutralizing/therapeutic use , Betacoronavirus/immunology , Betacoronavirus/pathogenicity , Caco-2 Cells , Gene Expression Regulation , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Humans , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/metabolism , SARS-CoV-2 , Signal Transduction , Viral Proteins/antagonists & inhibitors , Viral Proteins/metabolism , Virus Replication/drug effects
3.
PLoS Biol ; 19(1): e3001029, 2021 01.
Article in English | MEDLINE | ID: mdl-33395410

ABSTRACT

Endosomal trafficking of receptors and associated proteins plays a critical role in signal processing. Until recently, it was thought that trafficking was shut down during cell division. Thus, remarkably, the regulation of trafficking during division remains poorly characterized. Here we delineate the role of mitotic kinases in receptor trafficking during asymmetric division. Targeted perturbations reveal that Cyclin-dependent Kinase 1 (CDK1) and Aurora Kinase promote storage of Fibroblast Growth Factor Receptors (FGFRs) by suppressing endosomal degradation and recycling pathways. As cells progress through metaphase, loss of CDK1 activity permits differential degradation and targeted recycling of stored receptors, leading to asymmetric induction. Mitotic receptor storage, as delineated in this study, may facilitate rapid reestablishment of signaling competence in nascent daughter cells. However, mutations that limit or enhance the release of stored signaling components could alter daughter cell fate or behavior thereby promoting oncogenesis.


Subject(s)
Aurora Kinases/physiology , CDC2 Protein Kinase/physiology , Mitosis/physiology , Receptors, Fibroblast Growth Factor/metabolism , Animals , Animals, Genetically Modified , Aurora Kinases/genetics , CDC2 Protein Kinase/genetics , Cell Cycle Proteins/metabolism , Ciona intestinalis/embryology , Ciona intestinalis/genetics , Embryo, Nonmammalian , Mitosis/genetics , Protein Transport/genetics , Receptors, Fibroblast Growth Factor/genetics , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Signal Transduction/genetics , Tissue Distribution/genetics
4.
Anticancer Drugs ; 35(1): 76-80, 2024 01 01.
Article in English | MEDLINE | ID: mdl-37067984

ABSTRACT

Lung cancer is the malignancy with the highest morbidity and mortality worldwide. Approximately 60% of non-small cell lung cancer (NSCLC) presents driver alterations most of which are targetable. Nowadays, limited clinical data are available regarding the efficacy of epithelial growth factor receptor (EGFR) tyrosine kinase inhibitors in patients with NSCLC harboring uncommon EGFR mutations, considering their heterogeneity. Herein, we report a rare case of EGFR-mutated lung adenocarcinoma which has developed into squamous cell carcinoma with uncommon EGFR (Ex18) compound mutations and phosphatidylinositol 3-kinase mutation receiving afatinib at the forefront.


Subject(s)
Adenocarcinoma of Lung , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , ErbB Receptors/genetics , Mutation , Adenocarcinoma of Lung/drug therapy , Adenocarcinoma of Lung/genetics , Receptors, Growth Factor/genetics
5.
Toxicol Appl Pharmacol ; 437: 115886, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35041852

ABSTRACT

Many small molecule kinase inhibitors (SMKIs), used predominantly in cancer therapy, have been implicated in serious clinical cardiac adverse events, which means that traditional preclinical drug development assays were not sufficient for identifying these cardiac liabilities. To improve clinical cardiac safety predictions, the effects of SMKIs targeting many different signaling pathways were studied using human pluripotent stem cell derived cardiomyocytes (hPSC-CMs) in combined assays designed for the detection of both electrophysiological (proarrhythmic) and non-electrophysiological (non-proarrhythmic) drug-induced cardiotoxicity. Several microplate-based assays were used to quantitate cell death, apoptosis, mitochondrial damage, energy depletion, and oxidative stress as mechanism-based non-electrophysiological cardiomyocyte toxicities. Microelectrode arrays (MEA) were used to quantitate in vitro arrhythmic events (iAEs), field potential duration (FPD) prolongation, and spike amplitude suppression (SAS) as electrophysiological effects. To enhance the clinical relevance, SMKI-induced cardiotoxicities were compared by converting drug concentrations into multiples of reported clinical maximum therapeutic plasma concentration, "FoldCmax", for each assay. The results support the conclusion that the combination of the hPSC-CM based electrophysiological and non-electrophysiological assays have significantly more predictive value than either assay alone and significantly more than the current FDA-recommended hERG assay. In addition, the combination of these assays provided mechanistic information relevant to cardiomyocyte toxicities, thus providing valuable information on potential drug-induced cardiotoxicities early in drug development prior to animal and clinical testing. We believe that this early information will be helpful to guide the development of safer and more cost-effective drugs.


Subject(s)
Myocytes, Cardiac/drug effects , Pluripotent Stem Cells/physiology , Protein Kinase Inhibitors/pharmacology , Cell Differentiation , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Protein Kinase Inhibitors/chemistry , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism
6.
Proc Natl Acad Sci U S A ; 116(28): 13729-13737, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31213534

ABSTRACT

Cell surface carbohydrates, termed "glycans," are ubiquitous posttranslational effectors that can tune cancer progression. Often aberrantly displayed or found at atypical levels on cancer cells, glycans can impact essentially all progressive steps, from malignant transformation to metastases formation. Glycans are structural entities that can directly bind promalignant glycan-binding proteins and help elicit optimal receptor-ligand activity of growth factor receptors, integrins, integrin ligands, lectins, and other type-1 transmembrane proteins. Because glycans play an integral role in a cancer cell's malignant activity and are frequently uniquely expressed, preclinical studies on the suitability of glycans as anticancer therapeutic targets and their promise as biomarkers of disease progression continue to intensify. While sialylation and fucosylation have predominated the focus of cancer-associated glycan modifications, the emergence of blood group I antigens (or I-branched glycans) as key cell surface moieties capable of modulating cancer virulence has reenergized investigations into the role of the glycome in malignant progression. I-branched glycans catalyzed principally by the I-branching enzyme GCNT2 are now indicated in several malignancies. In this Perspective, the putative role of GCNT2/I-branching in cancer progression is discussed, including exciting insights on how I-branches can potentially antagonize the cancer-promoting activity of ß-galactose-binding galectins.


Subject(s)
Galectins/genetics , N-Acetylhexosaminyltransferases/genetics , Neoplasms/genetics , Polysaccharides/genetics , Carbohydrates/chemistry , Carbohydrates/genetics , Carrier Proteins/genetics , Disease Progression , Glycosylation , Humans , Integrins/genetics , Lectins/genetics , Neoplasms/metabolism , Neoplasms/pathology , Polysaccharides/metabolism , Receptors, Growth Factor/genetics , Signal Transduction
7.
Mol Pharmacol ; 100(5): 491-501, 2021 11.
Article in English | MEDLINE | ID: mdl-34470776

ABSTRACT

The neurotrophin growth factors bind and activate two types of cell surface receptors: the tropomyosin receptor kinase (Trk) family and p75. TrkA, TrkB, and TrkC are bound preferentially by nerve growth factor, brain-derived neurotrophic factor, and neurotrophin 3 (NT3), respectively, to activate neuroprotective signals. The p75 receptors are activated by all neurotrophins, and paradoxically in neurodegenerative disease p75 is upregulated and mediates neurotoxic signals. To test neuroprotection strategies, we engineered NT3 to broadly activate Trk receptors (mutant D) or to reduce p75 binding (mutant RK). We also combined these features in a molecule that activates TrkA, TrkB, and TrkC but has reduced p75 binding (mutant DRK). In neurodegenerative disease mouse models in vivo, the DRK protein is a superior therapeutic agent compared with mutant D, mutant RK, and wild-type neurotrophins and protects a broader range of stressed neurons. This work rationalizes a therapeutic strategy based on the biology of each type of receptor, avoiding activation of p75 toxicity while broadly activating neuroprotection in stressed neuronal populations expressing different Trk receptors. SIGNIFICANCE STATEMENT: The neurotrophins nerve growth factor, brain-derived neurotrophic factor, and neurotrophin 3 each can activate a tropomyosin receptor kinase (Trk) A, TrkB, or TrkC receptor, respectively, and all can activate a p75 receptor. Trks and p75 mediate opposite signals. We report the engineering of a protein that activates all Trks, combined with low p75 binding, as an effective therapeutic agent in vivo.


Subject(s)
Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Neuroprotection/physiology , Protein Engineering/methods , Receptor, trkA/metabolism , Receptors, Growth Factor/metabolism , Animals , Axotomy/adverse effects , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/genetics , Diabetic Neuropathies/metabolism , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Nerve Growth Factors/administration & dosage , Nerve Growth Factors/genetics , Nerve Tissue Proteins/genetics , Neuroprotection/drug effects , Optic Nerve/drug effects , Optic Nerve/metabolism , Receptor, trkA/genetics , Receptors, Growth Factor/genetics
8.
Biochem J ; 477(20): 4053-4070, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33043964

ABSTRACT

The tropomyosin-related kinase (Trk) family consists of three receptor tyrosine kinases (RTKs) called TrkA, TrkB, and TrkC. These RTKs are regulated by the neurotrophins, a class of secreted growth factors responsible for the development and function of neurons. The Trks share a high degree of homology and utilize overlapping signaling pathways, yet their signaling is associated with starkly different outcomes in certain cancers. For example, in neuroblastoma, TrkA expression and signaling correlates with a favorable prognosis, whereas TrkB is associated with poor prognoses. To begin to understand how activation of the different Trks can lead to such distinct cellular outcomes, we investigated differences in kinase activity and duration of autophosphorylation for the TrkA and TrkB tyrosine kinase domains (TKDs). We find that the TrkA TKD has a catalytic efficiency that is ∼2-fold higher than that of TrkB, and becomes autophosphorylated in vitro more rapidly than the TrkB TKD. Studies with mutated TKD variants suggest that a crystallographic dimer seen in many TrkA (but not TrkB) TKD crystal structures, which involves the kinase-insert domain, may contribute to this enhanced TrkA autophosphorylation. Consistent with previous studies showing that cellular context determines whether TrkB signaling is sustained (promoting differentiation) or transient (promoting proliferation), we also find that TrkB signaling can be made more transient in PC12 cells by suppressing levels of p75NTR. Our findings shed new light on potential differences between TrkA and TrkB signaling, and suggest that subtle differences in signaling dynamics can lead to substantial shifts in the cellular outcome.


Subject(s)
Neuroblastoma/metabolism , Receptor, trkA/metabolism , Receptor, trkB/metabolism , Signal Transduction/genetics , Animals , Brain-Derived Neurotrophic Factor/pharmacology , Catalytic Domain , Cell Differentiation/genetics , Cell Proliferation/genetics , Gene Knockdown Techniques , Kinetics , Mutation , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroblastoma/enzymology , Neuroblastoma/genetics , PC12 Cells , Phosphorylation , Protein Domains , RNA, Small Interfering , Rats , Receptor, trkA/chemistry , Receptor, trkA/genetics , Receptor, trkB/chemistry , Receptor, trkB/genetics , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Recombinant Proteins , Signal Transduction/drug effects
9.
Reprod Domest Anim ; 56(2): 351-359, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33259113

ABSTRACT

Ovarian response of collared peccaries (Pecari tajacu), after hormonal stimulation with gonadotropin association (eCG/hCG), was accessed by both gene expression and follicular development. Thus, collared peccaries (n = 8) were treated with the dose used for sows (swine dose, SWD) or with dose adjusted for peccary's weight (allometric dose, ALD). The gene expression of receptors was evaluated for both gonadotropins (FSHR and LHCGR) and growth factors (proteins codified by TGFßR-1, BMPR1-A and BMPR2 genes) in antral follicles, cortex and corpora haemorrhagica (CH). Five days after gonadotropin injection, all females presented CH. The ovulation rate was similar (p > .05) between SWD (4.00 ± 1.17) and ALD (2.50 ± 0.43) group. The total number of follicles per animal and amounts of small (<3 mm), medium (3-5 mm) and large (>5 mm) follicles was similar among groups. However, SWD produced large follicles heavier than ALD group, as accessed by weight of follicular wall biopsies. Ovarian follicles expressed both gonadotropin and growth factor receptors at levels which are independent from gonadotropin dose. In conclusion, the two gonadotropin doses (SWD and ALD) can be used for ovarian stimulation of collared peccary. Additionally, FSH and growth factors (TGFßR-1, BMPR1-A and BMPR2) receptors are more expressed in the early follicle development, while LH receptor seems to be more important in the final of follicular growth.


Subject(s)
Artiodactyla/physiology , Chorionic Gonadotropin/pharmacology , Ovary/drug effects , Animals , Body Weight , Chorionic Gonadotropin/administration & dosage , Female , Ovarian Follicle/drug effects , Ovulation/drug effects , Receptors, Gonadotropin/genetics , Receptors, Gonadotropin/metabolism , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism
10.
Int J Mol Sci ; 21(19)2020 10 01.
Article in English | MEDLINE | ID: mdl-33019660

ABSTRACT

The carotid body may undergo plasticity changes during development/ageing and in response to environmental (hypoxia and hyperoxia), metabolic, and inflammatory stimuli. The different cell types of the carotid body express a wide series of growth factors and corresponding receptors, which play a role in the modulation of carotid body function and plasticity. In particular, type I cells express nerve growth factor, brain-derived neurotrophic factor, neurotrophin 3, glial cell line-derived neurotrophic factor, ciliary neurotrophic factor, insulin-like-growth factor-I and -II, basic fibroblast growth factor, epidermal growth factor, transforming growth factor-α and -ß, interleukin-1ß and -6, tumor necrosis factor-α, vascular endothelial growth factor, and endothelin-1. Many specific growth factor receptors have been identified in type I cells, indicating autocrine/paracrine effects. Type II cells may also produce growth factors and express corresponding receptors. Future research will have to consider growth factors in further experimental models of cardiovascular, metabolic, and inflammatory diseases and in human (normal and pathologic) samples. From a methodological point of view, microarray and/or proteomic approaches would permit contemporary analyses of large groups of growth factors. The eventual identification of physical interactions between receptors of different growth factors and/or neuromodulators could also add insights regarding functional interactions between different trophic mechanisms.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Carotid Body/metabolism , Hyperoxia/genetics , Hypoxia/genetics , Nerve Growth Factor/genetics , Receptors, Growth Factor/genetics , Animals , Brain-Derived Neurotrophic Factor/metabolism , Carotid Body/cytology , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/metabolism , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Gene Expression Regulation , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Humans , Hyperoxia/metabolism , Hyperoxia/pathology , Hypoxia/metabolism , Hypoxia/pathology , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Nerve Growth Factor/metabolism , Neurotrophin 3/genetics , Neurotrophin 3/metabolism , Receptors, Growth Factor/metabolism , Transforming Growth Factor alpha/genetics , Transforming Growth Factor alpha/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
11.
J Stroke Cerebrovasc Dis ; 29(12): 105316, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32992173

ABSTRACT

OBJECTIVE: Pharmacological inhibition of GABAergic synapses could represent a potent neuromodulation strategy to activate hippocampal neurons and increase neurotrophic factor gene expression, thus exerting a beneficial effect on post-stroke cognitive impairment (PSCI). The objective of this study was to assess the effects of low-level inhibition of GABAergic synapses on hippocampal gene expressions related to neuroplasticity using the middle cerebral artery occlusion surgery (MCAO) ischemic stroke rat model. METHODS: The animals were randomly assigned to three experimental groups-(1) a sham operated group (SHAM), (2) a control group (CON), and (3) a bicuculline group (BIC). MCAO was performed in the CON and BIC groups. A non-epileptic dose of bicuculline (0.25 mg/kg) was intraperitoneally administered every day for two weeks, starting three days after surgery, to the rats in the BIC group. The mRNA expression of brain-derived neurotrophic factor (BDNF), tropomyosin-related kinase B (TrkB), in relation to neurotrophic intracellular signal, p75, in relation to apoptosis, and synaptophysin (SYP) and PSD-95, synaptic markers, were assessed in the hippocampus ipsilateral to the ischemic site. RESULTS: MCAO increased the gene expression of TrkB. Furthermore, MCAO plus bicuculline administration increased the expression ratio of TrkB to p75 and SYP gene expression. CONCLUSION: Therefore, this study showed that administration of bicuculline after stroke beneficially modulated the expression of crucial genes for neuroplasticity, including BDNF receptors and SYP, in the ipsilateral hippocampus, suggesting that low-level inhibition of GABAergic synapses could lead to beneficial neuromodulation in the hippocampus after stroke.


Subject(s)
Bicuculline/pharmacology , GABA-A Receptor Antagonists/pharmacology , GABAergic Neurons/drug effects , Hippocampus/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/genetics , Neural Inhibition/drug effects , Neuronal Plasticity/drug effects , Synaptic Transmission/drug effects , Animals , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , Disks Large Homolog 4 Protein/genetics , Disks Large Homolog 4 Protein/metabolism , GABAergic Neurons/metabolism , GABAergic Neurons/pathology , Gene Expression Regulation , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Infarction, Middle Cerebral Artery/metabolism , Infarction, Middle Cerebral Artery/physiopathology , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Rats, Sprague-Dawley , Receptor, trkB/genetics , Receptor, trkB/metabolism , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Synaptophysin/genetics , Synaptophysin/metabolism
12.
Prog Mol Subcell Biol ; 57: 181-202, 2018.
Article in English | MEDLINE | ID: mdl-30097776

ABSTRACT

Growth factor receptors play a variety of roles during embryonic development and in adult homeostasis. These receptors are activated repeatedly in different cellular contexts and with different cellular outcomes. This begs the question as to how cells in a particular developmental, spatial and temporal context, or in adult tissue, interpret signalling by growth factor receptors in order to deliver qualitatively different signalling outputs. One mechanism by which this could occur is via endocytic regulation. The original paradigm for the role of endocytosis in growth factor receptor signalling was that receptor uptake has a quantitative role in signalling by reducing the number of cell surface receptors available for activation and targeting activated receptors for degradation. However, a range of studies over the last several years, in many different experimental systems, has demonstrated an additional qualitative role for endocytic trafficking in receptor signalling, with specific outcomes depending on the location of the signalling complex. Confinement of receptors within endosomes can spatially regulate signalling, facilitating specific protein interactions or post-translational modifications that alter throughout the trafficking process. Therefore, endocytosis does not simply regulate cell surface expression, but tightly controls protein interactions and function to produce distinct outcomes.


Subject(s)
Endocytosis/genetics , Endosomes/genetics , Metabolic Networks and Pathways/genetics , Receptors, Growth Factor/genetics , Animals , Cell Membrane/genetics , Humans , Receptors, Cell Surface/genetics
13.
Genome Res ; 25(10): 1521-35, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26430163

ABSTRACT

Sporadic breast cancer (SBC) is a common disease without robust means of early risk prediction in the population. We studied 282 females with SBC, focusing on copy number aberrations in cancer-free breast tissue (uninvolved margin, UM) outside the primary tumor (PT). In total, 1162 UMs (1-14 per breast) were studied. Comparative analysis between UM(s), PT(s), and blood/skin from the same patient as a control is the core of the study design. We identified 108 patients with at least one aberrant UM, representing 38.3% of cases. Gains in gene copy number were the principal type of mutations in microscopically normal breast cells, suggesting that oncogenic activation of genes via increased gene copy number is a predominant mechanism for initiation of SBC pathogenesis. The gain of ERBB2, with overexpression of HER2 protein, was the most common aberration in normal cells. Five additional growth factor receptor genes (EGFR, FGFR1, IGF1R, LIFR, and NGFR) also showed recurrent gains, and these were occasionally present in combination with the gain of ERBB2. All the aberrations found in the normal breast cells were previously described in cancer literature, suggesting their causative, driving role in pathogenesis of SBC. We demonstrate that analysis of normal cells from cancer patients leads to identification of signatures that may increase risk of SBC and our results could influence the choice of surgical intervention to remove all predisposing cells. Early detection of copy number gains suggesting a predisposition toward cancer development, long before detectable tumors are formed, is a key to the anticipated shift into a preventive paradigm of personalized medicine for breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast/anatomy & histology , Mutation , Adult , Aged , Aged, 80 and over , Breast/pathology , Breast Neoplasms/pathology , Cohort Studies , DNA Mutational Analysis , Female , Gene Dosage , Genes, erbB-2 , Genetic Predisposition to Disease , Genotype , Humans , Middle Aged , Receptor, ErbB-2/genetics , Receptors, Growth Factor/genetics , Risk Factors
14.
Exp Eye Res ; 173: 129-137, 2018 08.
Article in English | MEDLINE | ID: mdl-29753729

ABSTRACT

Emerging therapies have begun to evaluate the abilities of Müller glial cells (MGCs) to protect and/or regenerate neurons following retina injury. The migration of donor cells is central to many reparative strategies, where cells must achieve appropriate positioning to facilitate localized repair. Although chemical cues have been implicated in the MGC migratory responses of numerous retinopathies, MGC-based therapies have yet to explore the extent to which external biochemical stimuli can direct MGC behavior. The current study uses a microfluidics-based assay to evaluate the migration of cultured rMC-1 cells (as model MGC) in response to quantitatively-controlled microenvironments of signaling factors implicated in retinal regeneration: basic Fibroblast Growth factor (bFGF or FGF2); Fibroblast Growth factor 8 (FGF8); Vascular Endothelial Growth Factor (VEGF); and Epidermal Growth Factor (EGF). Findings indicate that rMC-1 cells exhibited minimal motility in response to FGF2, FGF8 and VEGF, but highly-directional migration in response to EGF. Further, the responses were blocked by inhibitors of EGF-R and of the MAPK signaling pathway. Significantly, microfluidics data demonstrate that changes in the EGF gradient (i.e. change in EGF concentration over distance) resulted in the directional chemotactic migration of the cells. By contrast, small increases in EGF concentration, alone, resulted in non-directional cell motility, or chemokinesis. This microfluidics-enhanced approach, incorporating the ability both to modulate and asses the responses of motile donor cells to a range of potential chemotactic stimuli, can be applied to potential donor cell populations obtained directly from human specimens, and readily expanded to incorporate drug-eluting biomaterials and combinations of desired ligands.


Subject(s)
Chemotaxis/physiology , Ependymoglial Cells/physiology , Animals , Cell Proliferation/physiology , Cells, Cultured , Cellular Microenvironment , Ependymoglial Cells/drug effects , Epidermal Growth Factor/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Erlotinib Hydrochloride/pharmacology , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 8/pharmacology , Glial Fibrillary Acidic Protein/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Microfluidic Analytical Techniques , Nestin/metabolism , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction , Receptors, Growth Factor/genetics , Vascular Endothelial Growth Factor A/pharmacology
15.
Proc Natl Acad Sci U S A ; 112(9): E937-46, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25713130

ABSTRACT

Environmental cues are transmitted to the interior of the cell via a complex network of signaling hubs. Receptor tyrosine kinases (RTKs) and trimeric G proteins are two such major signaling hubs in eukaryotes. Conventionally, canonical signal transduction via trimeric G proteins is thought to be triggered exclusively by G protein-coupled receptors. Here we used molecular engineering to develop modular fluorescent biosensors that exploit the remarkable specificity of bimolecular recognition, i.e., of both G proteins and RTKs, and reveal the workings of a novel platform for activation of G proteins by RTKs in single living cells. Comprised of the unique modular makeup of guanidine exchange factor Gα-interacting vesicle-associated protein (GIV)/girdin, a guanidine exchange factor that links G proteins to a variety of RTKs, these biosensors provide direct evidence that RTK-GIV-Gαi ternary complexes are formed in living cells and that Gαi is transactivated within minutes after growth factor stimulation at the plasma membrane. Thus, GIV-derived biosensors provide a versatile strategy for visualizing, monitoring, and manipulating the dynamic association of Gαi with RTKs for noncanonical transactivation of G proteins in cells and illuminate a fundamental signaling event regulated by GIV during diverse cellular processes and pathophysiologic states.


Subject(s)
Biosensing Techniques/methods , Fluorescence Resonance Energy Transfer/methods , GTP-Binding Proteins , Receptor Protein-Tyrosine Kinases , Receptors, Growth Factor , Signal Transduction , Animals , COS Cells , Chlorocebus aethiops , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
16.
Gynecol Obstet Invest ; 83(4): 338-348, 2018.
Article in English | MEDLINE | ID: mdl-29241203

ABSTRACT

AIM: The aim is to identify complex pathogenesis of breast cancer subtypes and disclose the whole landscape of altered transcriptional activities in these cancers. METHODS: We downloaded raw expression data from public database, and performed transcriptome analysis of 8 estrogen receptor-positive (ER+) breast cancer tissue samples, 8 human epithelial growth factor receptor 2-positive (HER2+) breast cancer tissue samples, and 3 normal breast tissues by identification, functional annotation, and prediction of upstream regulators and cell-surface biomarkers of differentially expressed genes (DEGs). RESULTS: We identified over 5,000 DEGs in each of ER+ and HER2+ breast cancers compared to normal tissues. Functional enrichment analysis of shared DEGs indicated significant changes in the regulation of immune -systems in the 2 subtypes. We further identified 1,871 DEGs between the 2 subtypes and disclosed great tumor heterogeneity. We identified 533 shared upregulated genes and predicted 17 upstream transcription factors, as well as identified differentially expressed cell-surface biomarkers for distinguishing our ER+ and HER2+ breast cancers. Further analysis also highlighted the limitation of the usage of HER2 alone in breast cancer classification. CONCLUSION: Our findings in ER+ and HER2+ breast cancers provided novel insights into heterogeneous transcriptional activities underlying complex mechanisms of oncogenesis in breast cancers.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Gene Expression Profiling/methods , Receptor, ErbB-2/genetics , Receptors, Estrogen/genetics , Receptors, Growth Factor/genetics , Databases, Genetic , Female , Humans , Sequence Analysis, RNA
17.
Acta Chir Belg ; 118(3): 161-166, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29065778

ABSTRACT

INTRODUCTION: Anastomotic leakage after gastrointestinal surgery is a significant cause of morbidity and mortality. Esophagogastric and colorectal anastomoses are vulnerable to leakage. Extended knowledge of growth factors and their receptors is needed to understand anatomic healing. METHODS: The expression pattern of vascular growth factor receptor (VEGFR1-3), epidermal growth factor receptor (EGFR), platelet-derived growth factor receptor (PDGFRα/ß) and keratinocyte growth factor receptor (KGFR) were analyzed by semiquantitative-PCR in the rat intestinal tract and in esophagogastric anastomosis 5d after surgery. RESULTS: VEGFR1, VEGFR2, EGFR, KGFR and PDGFRα expression was observed throughout the intestinal tract including esophagus, stomach, small bowl and colon. VEGFR3 was not found in gastric samples and PDGFRß expression was not detected in the small bowl. Semiquantitative analyses of the VEGFR1, PDGFRα and EGFR expression in esophagogastric anastomotic tissues revealed a 2-fold upregulation of the VEGFR1 in gastric samples, while no change was observed in the esophageal anastomotic side. CONCLUSION: Our results revealed a distinct expression pattern of the investigated growth factor receptors in rat intestinal tract. Showing higher expression levels of growth factor receptors at the gastric anastomotic tissue at the fifth postoperative day suggests a different contribution of the gastric and the esophageal side to the anastomotic healing.


Subject(s)
Anastomotic Leak/diagnosis , Receptors, Growth Factor/genetics , Up-Regulation , Vascular Endothelial Growth Factor Receptor-1/genetics , Wound Healing/genetics , Analysis of Variance , Anastomosis, Surgical/adverse effects , Anastomosis, Surgical/methods , Animals , Biopsy, Needle , Digestive System Surgical Procedures/adverse effects , Digestive System Surgical Procedures/methods , Disease Models, Animal , Esophagogastric Junction/surgery , Gene Expression Regulation , Immunohistochemistry , Multivariate Analysis , RNA, Messenger/genetics , Random Allocation , Rats , Rats, Inbred BN , Real-Time Polymerase Chain Reaction , Wound Healing/physiology
18.
EMBO J ; 32(11): 1613-25, 2013 May 29.
Article in English | MEDLINE | ID: mdl-23591430

ABSTRACT

The sympathetic nervous system relies on distinct populations of neurons that use noradrenaline or acetylcholine as neurotransmitter. We show that fating of the sympathetic lineage at early stages results in hybrid precursors from which, genetic cell-lineage tracing reveals, all types progressively emerge by principal mechanisms of maintenance, repression and induction of phenotypes. The homeobox transcription factor HMX1 represses Tlx3 and Ret, induces TrkA and maintains tyrosine hydroxylase (Th) expression in precursors, thus driving segregation of the noradrenergic sympathetic fate. Cholinergic sympathetic neurons develop through cross-regulatory interactions between TRKC and RET in precursors, which lead to Hmx1 repression and sustained Tlx3 expression, thereby resulting in failure of TrkA induction and loss of maintenance of Th expression. Our results provide direct evidence for a model in which diversification of noradrenergic and cholinergic sympathetic neurons is based on a principle of cross-repressive functions in which the specific cell fates are directed by an active suppression of the expression of transcription factors and receptors that direct the alternative fate.


Subject(s)
Cell Differentiation , Cholinergic Neurons/cytology , Homeodomain Proteins/metabolism , Receptors, Growth Factor/metabolism , Transcription Factors/metabolism , Tyrosine 3-Monooxygenase/genetics , Acetylcholine/metabolism , Adrenergic alpha-Agonists/metabolism , Animals , Cholinergic Agonists/metabolism , Cholinergic Neurons/physiology , Chromosomes, Artificial, Bacterial , Embryo, Mammalian , Female , Gene Expression Regulation, Developmental , Gene Library , Homeodomain Proteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Norepinephrine/metabolism , Phenotype , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Receptor, trkC/genetics , Receptor, trkC/metabolism , Receptors, Growth Factor/genetics , Sympathetic Nervous System/cytology , Sympathetic Nervous System/physiology , Transcription Factors/genetics , Tyrosine 3-Monooxygenase/metabolism
19.
Amino Acids ; 49(3): 425-439, 2017 03.
Article in English | MEDLINE | ID: mdl-27562794

ABSTRACT

The ability of cancer cells to metastasize represents the most devastating feature of cancer. Currently, there are no specific biomarkers or therapeutic targets that can be used to predict the risk or to treat metastatic cancer. Many recent reports have demonstrated elevated expression of transglutaminase 2 (TG2) in multiple drug-resistant and metastatic cancer cells. TG2 is a multifunctional protein mostly known for catalyzing Ca2+-dependent -acyl transferase reaction to form protein crosslinks. Besides this transamidase activity, many Ca2+-independent and non-enzymatic activities of TG2 have been identified. Both, the enzymatic and non-enzymatic activities of TG2 have been implicated in diverse pathophysiological processes such as wound healing, cell growth, cell survival, extracellular matrix modification, apoptosis, and autophagy. Tumors have been frequently referred to as 'wounds that never heal'. Based on the observation that TG2 plays an important role in wound healing and inflammation is known to facilitate cancer growth and progression, we discuss the evidence that TG2 can reprogram inflammatory signaling networks that play fundamental roles in cancer progression. TG2-regulated signaling bestows on cancer cells the ability to proliferate, to resist cell death, to invade, to reprogram glucose metabolism and to metastasize, the attributes that are considered important hallmarks of cancer. Therefore, inhibiting TG2 may offer a novel therapeutic approach for managing and treatment of metastatic cancer. Strategies to inhibit TG2-regulated pathways will also be discussed.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , GTP-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Neoplasms/drug therapy , Signal Transduction , Transglutaminases/genetics , Apoptosis/drug effects , Autophagy/drug effects , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Cell Proliferation , Cell Survival/drug effects , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/metabolism , Humans , Molecular Targeted Therapy , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Metastasis , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Protein Glutamine gamma Glutamyltransferase 2 , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Transglutaminases/antagonists & inhibitors , Transglutaminases/metabolism , Wound Healing/genetics
20.
Can J Physiol Pharmacol ; 95(8): 945-953, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28704617

ABSTRACT

We previously showed that augmented levels of endogenous angiotensin II (AngII) contribute to vascular smooth muscle cell (VSMC) hypertrophy through the transactivation of growth factor receptors in spontaneously hypertensive rats. Resveratrol (RV), a polyphenolic component of red wine, has also been shown to attenuate AngII-evoked VSMC hypertrophy; however, the molecular mechanism mediating this response is obscure. The present study was therefore undertaken to examine whether RV could prevent AngII-induced VSMC hypertrophy through the transactivation of growth factor receptor and associated signaling pathways. AngII treatment of VSMC enhanced the protein synthesis that was attenuated towards control levels by RV pretreatment as well as by the inhibitors of NADPH oxidase, c-Src, and growth factor receptors. Furthermore, RV pretreatment also inhibited enhanced levels of superoxide anion, NADPH oxidase activity, increased expression of NADPH oxidase subunits, and phosphorylation of c-Src, EGF-R, PDGE-R, ERK1/2, and AKT1/2. In conclusion, these results indicate that RV attenuates AngII-induced VSMC hypertrophy through the inhibition of enhanced oxidative stress and activation of c-Src, growth factor receptors, and MAPK/AKT signaling. We suggest that RV could be used as a therapeutic agent in the treatment of vascular complications associated with hypertension and hypertrophy.


Subject(s)
Angiotensin II/pharmacology , Antihypertensive Agents/pharmacology , Muscle, Smooth, Vascular/pathology , Receptors, Growth Factor/genetics , Stilbenes/pharmacology , Transcriptional Activation/drug effects , Animals , CSK Tyrosine-Protein Kinase , Cell Line , Enzyme Activation/drug effects , Hypertrophy/chemically induced , Hypertrophy/genetics , Hypertrophy/metabolism , Hypertrophy/prevention & control , Male , Muscle, Smooth, Vascular/drug effects , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Resveratrol , Signal Transduction/drug effects , src-Family Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL