Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Mol Cell Proteomics ; 23(7): 100780, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38703893

ABSTRACT

New tools for cell signaling pathway inference from multi-omics data that are independent of previous knowledge are needed. Here, we propose a new de novo method, the de novo multi-omics pathway analysis (DMPA), to model and combine omics data into network modules and pathways. DMPA was validated with published omics data and was found accurate in discovering reported molecular associations in transcriptome, interactome, phosphoproteome, methylome, and metabolomics data, and signaling pathways in multi-omics data. DMPA was benchmarked against module discovery and multi-omics integration methods and outperformed previous methods in module and pathway discovery especially when applied to datasets of relatively low sample sizes. Transcription factor, kinase, subcellular location, and function prediction algorithms were devised for transcriptome, phosphoproteome, and interactome modules and pathways, respectively. To apply DMPA in a biologically relevant context, interactome, phosphoproteome, transcriptome, and proteome data were collected from analyses carried out using melanoma cells to address gamma-secretase cleavage-dependent signaling characteristics of the receptor tyrosine kinase TYRO3. The pathways modeled with DMPA reflected the predicted function and its direction in validation experiments.


Subject(s)
Proteomics , Signal Transduction , Humans , Proteomics/methods , Algorithms , Transcriptome , Metabolomics/methods , Computational Biology/methods , Proteome/metabolism , Phosphoproteins/metabolism , Multiomics
2.
EMBO Rep ; 24(5): e56689, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37009825

ABSTRACT

The growth factor Neuregulin-1 (NRG-1) regulates myocardial growth and is currently under clinical investigation as a treatment for heart failure. Here, we demonstrate in several in vitro and in vivo models that STAT5b mediates NRG-1/EBBB4-stimulated cardiomyocyte growth. Genetic and chemical disruption of the NRG-1/ERBB4 pathway reduces STAT5b activation and transcription of STAT5b target genes Igf1, Myc, and Cdkn1a in murine cardiomyocytes. Loss of Stat5b also ablates NRG-1-induced cardiomyocyte hypertrophy. Dynamin-2 is shown to control the cell surface localization of ERBB4 and chemical inhibition of Dynamin-2 downregulates STAT5b activation and cardiomyocyte hypertrophy. In zebrafish embryos, Stat5 is activated during NRG-1-induced hyperplastic myocardial growth, and chemical inhibition of the Nrg-1/Erbb4 pathway or Dynamin-2 leads to loss of myocardial growth and Stat5 activation. Moreover, CRISPR/Cas9-mediated knockdown of stat5b results in reduced myocardial growth and cardiac function. Finally, the NRG-1/ERBB4/STAT5b signaling pathway is differentially regulated at mRNA and protein levels in the myocardium of patients with pathological cardiac hypertrophy as compared to control human subjects, consistent with a role of the NRG-1/ERBB4/STAT5b pathway in myocardial growth.


Subject(s)
Dynamin II , Neuregulin-1 , Mice , Humans , Animals , Dynamin II/metabolism , Neuregulin-1/genetics , Neuregulin-1/metabolism , Neuregulin-1/pharmacology , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Zebrafish/metabolism , Receptor, ErbB-4/genetics , Receptor, ErbB-4/metabolism , Hypertrophy
3.
Biochem Biophys Res Commun ; 733: 150682, 2024 Nov 12.
Article in English | MEDLINE | ID: mdl-39276696

ABSTRACT

Global warming increases the risk of dangerous heat waves, which may have deleterious effects on humans and wildlife. Here, we have utilized zebrafish embryos as a model to analyze heat stress and effect of chemical compounds on responses to heat stress. The temperature adaptation limit of zebrafish embryos was 37 °C in behavioural test and 38 °C in cardiac test. Polyaromatic hydrocarbon phenanthrene completely blocked the behavioural adaptation to heat stress. Interestingly, the cardiotoxic effects of lapatinib, phenanthrene and paclitaxel were induced by heat stress. Taken together, our data indicates that motility and cardiac function of zebrafish embryos can be utilized as a model to analyze modulatory effects of compounds on heat stress.


Subject(s)
Embryo, Nonmammalian , Heat-Shock Response , Phenanthrenes , Zebrafish , Zebrafish/embryology , Animals , Heat-Shock Response/drug effects , Heat-Shock Response/physiology , Phenanthrenes/toxicity , Embryo, Nonmammalian/drug effects , Paclitaxel/toxicity , Paclitaxel/pharmacology , Heart/drug effects , Heart/embryology , Cardiotoxicity/etiology , Quinazolines/pharmacology , Quinazolines/toxicity
4.
J Biol Chem ; 295(33): 11559-11571, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32561640

ABSTRACT

ERBB4 is a member of the epidermal growth factor receptor (EGFR)/ERBB subfamily of receptor tyrosine kinases that regulates cellular processes including proliferation, migration, and survival. ERBB4 signaling is involved in embryogenesis and homeostasis of healthy adult tissues, but also in human pathologies such as cancer, neurological disorders, and cardiovascular diseases. Here, an MS-based analysis revealed the Vav guanine nucleotide exchange factor 3 (VAV3), an activator of Rho family GTPases, as a critical ERBB4-interacting protein in breast cancer cells. We confirmed the ERBB4-VAV3 interaction by targeted MS and coimmunoprecipitation experiments and further defined it by demonstrating that kinase activity and Tyr-1022 and Tyr-1162 of ERBB4, as well as the intact phosphotyrosine-interacting SH2 domain of VAV3, are necessary for this interaction. We found that ERBB4 stimulates tyrosine phosphorylation of the VAV3 activation domain, known to be required for guanine nucleotide exchange factor (GEF) activity of VAV proteins. In addition to VAV3, the other members of the VAV family, VAV1 and VAV2, also coprecipitated with ERBB4. Analyses of the effects of overexpression of dominant-negative VAV3 constructs or shRNA-mediated down-regulation of VAV3 expression in breast cancer cells indicated that active VAV3 is involved in ERBB4-stimulated cell migration. These results define the VAV GEFs as effectors of ERBB4 activity in a signaling pathway relevant for cancer cell migration.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , Proto-Oncogene Proteins c-vav/metabolism , Receptor, ErbB-4/metabolism , Animals , Breast Neoplasms/pathology , Female , HEK293 Cells , Humans , MCF-7 Cells , Mice , NIH 3T3 Cells , Protein Interaction Maps
5.
Circulation ; 139(22): 2570-2584, 2019 05 28.
Article in English | MEDLINE | ID: mdl-30922063

ABSTRACT

BACKGROUND: Heart failure, which is a major global health problem, is often preceded by pathological cardiac hypertrophy. The expansion of the cardiac vasculature, to maintain adequate supply of oxygen and nutrients, is a key determinant of whether the heart grows in a physiological compensated manner or a pathological decompensated manner. Bidirectional endothelial cell (EC)-cardiomyocyte (CMC) cross talk via cardiokine and angiocrine signaling plays an essential role in the regulation of cardiac growth and homeostasis. Currently, the mechanisms involved in the EC-CMC interaction are not fully understood, and very little is known about the EC-derived signals involved. Understanding how an excess of angiogenesis induces cardiac hypertrophy and how ECs regulate CMC homeostasis could provide novel therapeutic targets for heart failure. METHODS: Genetic mouse models were used to delete vascular endothelial growth factor (VEGF) receptors, adeno-associated viral vectors to transduce the myocardium, and pharmacological inhibitors to block VEGF and ErbB signaling in vivo. Cell culture experiments were used for mechanistic studies, and quantitative polymerase chain reaction, microarrays, ELISA, and immunohistochemistry were used to analyze the cardiac phenotypes. RESULTS: Both EC deletion of VEGF receptor (VEGFR)-1 and adeno-associated viral vector-mediated delivery of the VEGFR1-specific ligands VEGF-B or placental growth factor into the myocardium increased the coronary vasculature and induced CMC hypertrophy in adult mice. The resulting cardiac hypertrophy was physiological, as indicated by preserved cardiac function and exercise capacity and lack of pathological gene activation. These changes were mediated by increased VEGF signaling via endothelial VEGFR2, because the effects of VEGF-B and placental growth factor on both angiogenesis and CMC growth were fully inhibited by treatment with antibodies blocking VEGFR2 or by endothelial deletion of VEGFR2. To identify activated pathways downstream of VEGFR2, whole-genome transcriptomics and secretome analyses were performed, and the Notch and ErbB pathways were shown to be involved in transducing signals for EC-CMC cross talk in response to angiogenesis. Pharmacological or genetic blocking of ErbB signaling also inhibited part of the VEGF-B-induced effects in the heart. CONCLUSIONS: This study reveals that cross talk between the EC VEGFR2 and CMC ErbB signaling pathways coordinates CMC hypertrophy with angiogenesis, contributing to physiological cardiac growth.


Subject(s)
Cardiomegaly/metabolism , Endothelial Cells/metabolism , Myocytes, Cardiac/metabolism , Neovascularization, Physiologic , Paracrine Communication , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cells, Cultured , Disease Models, Animal , Endothelial Cells/pathology , ErbB Receptors/metabolism , Heparin-binding EGF-like Growth Factor/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Receptor Cross-Talk , Receptors, Notch/metabolism , Vascular Endothelial Growth Factor B/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics
6.
J Biol Chem ; 289(32): 22459-69, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-24966332

ABSTRACT

Conditional knock-out of Hif1a in the mouse mammary gland impairs lobuloalveolar differentiation during lactation. Here, we demonstrate that expression of ErbB4 was reduced in the lobulalveoli of mice with mammary gland-specific deletion of Hif1a. Erbb4 was not, however, a direct target gene for transcriptional regulation by HIF-1α in vitro. HIF-1α overexpression or HIF accumulating prolyl hydroxylase inhibitors reduced ErbB4 endocytosis, promoted transcriptional co-regulatory activity of ErbB4, and stimulated ErbB4-induced differentiation of mammary carcinoma cells. Consistently, RNA interference-mediated down-regulation of HIF-1α resulted in reduced ErbB4 protein amount and reduced mammary carcinoma cell differentiation. These findings indicate that HIF-1α is a physiologically relevant regulator of ErbB4 and that ErbB4 is involved in HIF-regulated differentiation of the mammary gland.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Receptor, ErbB-4/metabolism , Animals , Cell Differentiation , Cell Line, Tumor , Endocytosis , Female , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/deficiency , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lactation/genetics , Lactation/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Human/cytology , Mammary Glands, Human/growth & development , Mammary Glands, Human/metabolism , Mice , Mice, Knockout , Peptide Fragments/metabolism , Pregnancy , Signal Transduction
7.
J Biol Chem ; 287(27): 23216-26, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22584572

ABSTRACT

ErbB4 is a receptor tyrosine kinase implicated in the development and homeostasis of the heart, central nervous system, and mammary gland. Cleavable isoforms of ErbB4 release a soluble intracellular domain (ICD) that can translocate to the nucleus and function as a transcriptional coregulator. In search of regulatory mechanisms of ErbB4 ICD function, we identified PIAS3 as a novel interaction partner of ErbB4 ICD. In keeping with the small ubiquitin-like modifier (SUMO) E3 ligase function of protein inhibitor of activated STAT (PIAS) proteins, we showed that the ErbB4 ICD is modified by SUMO, and that PIAS3 stimulates the SUMOylation. Upon overexpression of PIAS3, the ErbB4 ICD generated from the full-length receptor accumulated into the nucleus in a manner that was dependent on the functional nuclear localization signal of ErbB4. In the nucleus, ErbB4 colocalized with PIAS3 and SUMO-1 in promyelocytic leukemia nuclear bodies, nuclear domains involved in regulation of transcription. Accordingly, PIAS3 overexpression had an effect on the transcriptional coregulatory activity of ErbB4, repressing its ability to coactivate transcription with Yes-associated protein. Finally, knockdown of PIAS3 with siRNA partially rescued the inhibitory effect of the ErbB4 ICD on differentiation of MDA-MB-468 breast cancer and HC11 mammary epithelial cells. Our findings illustrate that PIAS3 is a novel regulator of ErbB4 receptor tyrosine kinase, controlling its nuclear sequestration and function.


Subject(s)
Active Transport, Cell Nucleus/physiology , ErbB Receptors/metabolism , Molecular Chaperones/metabolism , Protein Inhibitors of Activated STAT/metabolism , Sumoylation/physiology , Animals , Breast Neoplasms , COS Cells , Cell Nucleus/metabolism , Chlorocebus aethiops , ErbB Receptors/chemistry , ErbB Receptors/genetics , Female , HEK293 Cells , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Molecular Chaperones/genetics , Nuclear Proteins/metabolism , Poly-ADP-Ribose Binding Proteins , Promyelocytic Leukemia Protein , Protein Inhibitors of Activated STAT/genetics , Protein Interaction Domains and Motifs/physiology , Protein Structure, Tertiary/physiology , RNA, Small Interfering/genetics , Receptor, ErbB-4 , Signal Transduction/physiology , Small Ubiquitin-Related Modifier Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism
8.
Nat Commun ; 13(1): 6953, 2022 11 14.
Article in English | MEDLINE | ID: mdl-36376313

ABSTRACT

The ErbB4 receptor isoforms JM-a and JM-b differ within their extracellular juxtamembrane (eJM) domains. Here, ErbB4 isoforms are used as a model to address the effect of structural variation in the eJM domain of receptor tyrosine kinases (RTK) on downstream signaling. A specific JM-a-like sequence motif is discovered, and its presence or absence (in JM-b-like RTKs) in the eJM domains of several RTKs is demonstrated to dictate selective STAT activation. STAT5a activation by RTKs including the JM-a like motif is shown to involve interaction with oligosaccharides of N-glycosylated cell surface proteins such as ß1 integrin, whereas STAT5b activation by JM-b is dependent on TYK2. ErbB4 JM-a- and JM-b-like RTKs are shown to associate with specific signaling complexes at different cell surface compartments using analyses of RTK interactomes and super-resolution imaging. These findings provide evidence for a conserved mechanism linking a ubiquitous extracellular motif in RTKs with selective intracellular STAT signaling.


Subject(s)
Receptor Protein-Tyrosine Kinases , Signal Transduction , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, ErbB-4/metabolism , Protein Isoforms/metabolism , Cell Membrane/metabolism , Phosphorylation
9.
Sci Rep ; 11(1): 16661, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34404849

ABSTRACT

The return of blood flow to ischemic heart after myocardial infarction causes ischemia-reperfusion injury. There is a clinical need for novel therapeutic targets to treat myocardial ischemia-reperfusion injury. Here we screened for targets for the treatment of ischemia-reperfusion injury using a combination of shRNA and drug library analyses in HL-1 mouse cardiomyocytes subjected to hypoxia and reoxygenation. The shRNA library included lentiviral constructs targeting 4625 genes and the drug library 689 chemical compounds approved by the Food and Drug Administration (FDA). Data were analyzed using protein-protein interaction and pathway analyses. EGFR inhibition was identified as a cardioprotective mechanism in both approaches. Inhibition of EGFR kinase activity with gefitinib improved cardiomyocyte viability in vitro. In addition, gefitinib preserved cardiac contractility in zebrafish embryos exposed to hypoxia-reoxygenation in vivo. These findings indicate that the EGFR inhibitor gefitinib is a potential candidate for further studies of repurposing the drug for the treatment of myocardial infarction.


Subject(s)
Cardiotonic Agents/pharmacology , Cell Hypoxia/drug effects , ErbB Receptors/antagonists & inhibitors , Gefitinib/pharmacology , Myocytes, Cardiac/drug effects , Protein Kinase Inhibitors/pharmacology , Animals , Cell Line , Disease Models, Animal , Drug Discovery , ErbB Receptors/genetics , Hypoxia/drug therapy , Hypoxia/genetics , Mice , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/genetics , Myocytes, Cardiac/metabolism , Zebrafish
10.
Sci Rep ; 8(1): 16579, 2018 11 08.
Article in English | MEDLINE | ID: mdl-30410004

ABSTRACT

Therapeutic protocols including EGFR antibodies in the context of oxaliplatin-based regimens have variable clinical effect in colorectal cancer. Here, we tested the effect of the EGFR antibody cetuximab in different sequential combinations with oxaliplatin on the growth of colorectal cancer cells in vitro and in vivo. Cetuximab reduced the efficacy of oxaliplatin when administered before oxaliplatin but provided additive effect when administered after oxaliplatin regardless of the KRAS or BRAF mutation status of the cells. Systemic gene expression and protein phosphorylation screens revealed alternatively activated pathways regulating apoptosis, cell cycle and DNA damage response. Functional assays indicated that cetuximab-induced arrest of the cells into the G1 phase of the cell cycle was associated with reduced responsiveness of the cells to subsequent treatment with oxaliplatin. In contrast, oxaliplatin-enhanced responsiveness to subsequent treatment with cetuximab was associated with increased apoptosis, inhibition of STAT3 activity and increased EGFR down-regulation. This preclinical study indicates that optimizing the sequence of administration may enhance the antitumor effect of combination therapy with EGFR antibodies and oxaliplatin.


Subject(s)
Cell Cycle/drug effects , Cetuximab/administration & dosage , Colorectal Neoplasms/drug therapy , DNA Repair/drug effects , Oxaliplatin/administration & dosage , Animals , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cetuximab/pharmacology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HT29 Cells , Humans , Mice , Mutation , Oxaliplatin/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , STAT3 Transcription Factor/metabolism , Xenograft Model Antitumor Assays
11.
Cell Cycle ; 10(16): 2647-57, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21811097

ABSTRACT

Alternative splicing is a central tool  of evolution that significantly increases the size of transcriptomes and generates functional specification. Within the human ERBB receptor gene family, only ERBB4 is known to produce functionally distinct isoforms as a result of alternative splicing. While ErbB4 signaling has been demonstrated to regulate cellular processes involved in embryogenesis, carcinogenesis and cardiovascular and psychiatric diseases, relatively little is known about the contribution of the individual isoforms in the different biological contexts. Here, we review recent findings as well as provide novel data about the distribution and functions of the ERBB4 splice variants. These observations represent an example of how minor alterations in the transcripts of a single gene can result in even antagonistic cellular responses. The observations also underline the significance of understanding the unique functions of isoforms of a potential drug target gene.


Subject(s)
Alternative Splicing , ErbB Receptors/metabolism , Animals , Cell Differentiation , ErbB Receptors/genetics , Humans , Neoplasms/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, ErbB-4 , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL