Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 149
Filter
1.
Endocrinology ; 165(5)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38578949

ABSTRACT

OBJECTIVES: Growth factor receptor bound protein 7 (GRB7) is a multidomain signaling adaptor. Members of the Grb7/10/14 family, specifically Gbrb10/14, have important roles in metabolism. We ablated the Grb7 gene in mice to examine its metabolic function. METHODS: Global ablation of Grb7 in FVB/NJ mice was generated. Growth, organ weight, food intake, and glucose homeostasis were measured. Insulin signaling was examined by Western blotting. Fat and lean body mass was measured by nuclear magnetic resonance, and body composition after fasting or high-fat diet was assessed. Energy expenditure was measured by indirect calorimetry. Expression of adiposity and lipid metabolism genes was measured by quantitative PCR. RESULTS: Grb7-null mice were viable, fertile, and without obvious phenotype. Grb7 ablation improved glycemic control and displayed sensitization to insulin signaling in the liver. Grb7-null females but not males had increased gonadal white adipose tissue mass. Following a 12-week high-fat diet, Grb7-null female mice gained fat body mass and developed relative insulin resistance. With fasting, there was less decrease in fat body mass in Grb7-null female mice. Female mice with Grb7 ablation had increased baseline food intake, less energy expenditure, and displayed a decrease in the expression of lipolysis and adipose browning genes in gonadal white adipose tissue by transcript and protein analysis. CONCLUSION: Our study suggests that Grb7 is a negative regulator of glycemic control. Our results reveal a role for Grb7 in female mice in the regulation of the visceral adipose tissue mass, a powerful predictor of metabolic dysfunction in obesity.


Subject(s)
Abdominal Fat , Energy Metabolism , GRB7 Adaptor Protein , Insulin , Mice, Knockout , Signal Transduction , Animals , Female , Male , Mice , Abdominal Fat/metabolism , Blood Glucose/metabolism , Body Composition/genetics , Diet, High-Fat , Energy Metabolism/genetics , GRB7 Adaptor Protein/genetics , GRB7 Adaptor Protein/metabolism , Insulin/metabolism , Insulin Resistance/genetics
2.
BMC Cancer ; 23(1): 1262, 2023 Dec 21.
Article in English | MEDLINE | ID: mdl-38129809

ABSTRACT

BACKGROUND: Gastric cancer is a clinically common tumor, showing an upward trend of both incidence and mortality. GRB7 has been identified as a vital regulator in tumor progression. This study aims to uncover the biological function of GRB7 in gastric cancer process. METHODS: immunohistochemical (IHC) staining using a tissue microarray (TMA), quantitative reverse transcription PCR (qRT-PCR) and Western blotting were performed to detect the expression of genes. Furthermore, gastric cancer cell lines AGS and MGC-803 were transfected with short hairpin RNAs against GRB7. The biological function of GRB7 in gastric cancer cells were examined by CCK-8, flow cytometry, wound healing and Transwell assays. Then, in vivo tumor formation assay was conducted to explore the effects of GRB7 on tumor growth. Finally, expression levels of proteins related to cell functions were determined by Western blotting. Coimmunoprecipitation (CoIP) assay was performed to assess the protein-protein interaction. RESULTS: GRB7 was up-regulated in gastric cancer tissues and cell lines, and its expression was inversely proportional to survival of gastric cancer patients. Moreover, GRB7 knockdown inhibited proliferative, migratory abilities, as well as promoted cell apoptosis in gastric cancer cells. Further study suggested that GRB7 silencing could suppress gastric cancer tumor growth in vivo. Furthermore, our study uncovered an important interaction between GRB7 and MyD88. Silencing MyD88 was observed to alleviate the malignant phenotypes promoted by GRB7 in gastric cancer cells. CONCLUSIONS: Together, this study provided evidence that GRB7 may be an effective molecular targets for the treatment of gastric cancer.


Subject(s)
Stomach Neoplasms , Humans , Cell Line, Tumor , Stomach Neoplasms/pathology , Myeloid Differentiation Factor 88/genetics , Cell Proliferation/genetics , RNA, Small Interfering , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , GRB7 Adaptor Protein/genetics , GRB7 Adaptor Protein/metabolism
3.
J Biol Chem ; 299(8): 104950, 2023 08.
Article in English | MEDLINE | ID: mdl-37354972

ABSTRACT

Xenopus oocytes are encompassed by a layer of follicular cells that contribute to oocyte growth and meiosis in relation to oocyte maturation. However, the effects of the interaction between follicular cells and the oocyte surface on meiotic processes are unclear. Here, we investigated Xenopus follicular cell function using oocyte signaling and heterologous-expressing capabilities. We found that oocytes deprotected from their surrounding layer of follicular cells and expressing the epidermal growth factor (EGF) receptor (EGFR) and the Grb7 adaptor undergo accelerated prophase I to metaphase II meiosis progression upon stimulation by EGF. This unusual maturation unravels atypical spindle formation but is rescued by inhibiting integrin ß1 or Grb7 binding to the EGFR. In addition, we determined that oocytes surrounded by their follicular cells expressing EGFR-Grb7 exhibit normal meiotic resumption. These oocytes are protected from abnormal meiotic spindle formation through the recruitment of O-GlcNAcylated Grb7, and OGT (O-GlcNAc transferase), the enzyme responsible for O-GlcNAcylation processes, in the integrin ß1-EGFR complex. Folliculated oocytes can be forced to adopt an abnormal phenotype and exclusive Grb7 Y338 and Y188 phosphorylation instead of O-GlcNAcylation under integrin activation. Furthermore, an O-GlcNAcylation increase (by inhibition of O-GlcNAcase), the glycosidase that removes O-GlcNAc moieties, or decrease (by inhibition of OGT) amplifies oocyte spindle defects when follicular cells are absent highlighting a control of the meiotic spindle by the OGT-O-GlcNAcase duo. In summary, our study provides further insight into the role of the follicular cell layer in oocyte meiosis progression.


Subject(s)
Epidermal Growth Factor , Integrin beta1 , Oocytes , Xenopus laevis , Animals , Acylation , Down-Regulation , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , GRB7 Adaptor Protein/metabolism , Integrin beta1/genetics , Integrin beta1/metabolism , Meiosis , Oocytes/cytology , Oocytes/growth & development , Oocytes/metabolism , Spindle Apparatus/metabolism , Xenopus laevis/metabolism
4.
Rev. colomb. cancerol ; 27(1): 80-90, 2023. graf, tab
Article in Spanish | LILACS, COLNAL | ID: biblio-1451954

ABSTRACT

Objetivo. Analizar las diferencias en la presentación de variables clínico-patológicas, de acuerdo con la expresión proteica de GRB7, en tumores HER2 positivos en mujeres colombianas con cáncer de mama invasivo, diagnosticado entre los años 2013 y 2015 en el Instituto Nacional de Cancerología E.S.E (INC). Métodos. Se incluyeron 158 pacientes con diagnóstico confirmado de cáncer de mama ductal invasivo. Se evaluó la expresión de los receptores hormonales (receptor de estrógeno (RE) y de progesterona (RP)), HER2, Ki67 y GRB7, mediante inmunohistoquímica (IHQ), y a partir de estos, se clasificaron los tumores en subtipos intrínsecos. Los análisis estadísticos incluyeron las pruebas de Chi-cuadrado/test exacto de Fisher para las variables categóricas, y la prueba U Mann Whitney/ Kruskal Wallis para las variables cuantitativas. Se evaluó la supervivencia global (SG) y libre de enfermedad (SLR) según la coexpresión de HER2/GRB7 usando el método de Kaplan-Meier y el test de log-rank. Resultados. La expresión de GRB7 se observó exclusivamente en tumores HER2-positivos (luminal B/HER2+ y HER2-enriquecidos: p<0,001). Los casos HER2+/GRB7+ mostraron una mayor expresión de Ki67 (40% vs. 27,5%, p=0,029), pero una tendencia a presentar un menor tamaño tumoral (30 mm vs. 51 mm, p=0,097), comparado con los tumores HER2+/GRB7-. No obstante, no se observaron diferencias en la supervivencia según la coexpresión de HER2/GRB7 (SG: p=0,6; SLR: p=0,07). Conclusiones. En nuestra muestra de estudio, la expresión de GRB7 en tumores HER2+ no se asoció con características clínico-patológicas de pronóstico desfavorable.


Objective: To analyze differences in the presentation of clinicopathological variables according to GRB7 protein expression in HER2-positive tumors in Colombian patients with invasive ductal breast carcinomas diagnosed between 2013 and 2015 at the Instituto Nacional de Cancerología (Bogotá, Colombia).Methods: A total of 158 breast cancer patients were included with a confirmed diagnosis of invasive ductal carcinoma. A single pathologist evaluated the protein expression of hormone receptors (estrogen (ER) and progesterone receptor (PR)), HER2, Ki67, and GRB7 by immunohistochemistry (IHC). The chi-square and Fisher's exact tests were used to assess differences between categorical variables, as well as the Mann-Whitney/Kruskal-Wallis U test for numerical variables. Overall (OS) and disease-free (DFS) survival were evaluated according to HER2/GRB7 co-expression using the Kaplan-Meier method and log-rank test.Results:GRB7 expression was observed exclusively in HER2-positive tumors (luminal B/HER2+ and HER2-enriched: p<0.001). HER2+/GRB7+ cases showed higher Ki67 expression (40% vs. 27.5%, p=0.029) and a tendency to present a smaller tumor (30 mm vs. 51 mm, p=0.097) compared to HER2+/GRB7- tumors. However, no differences in OS or DFS were observed by HER2/GRB7 co-expression (OS: p=0.6; DFS: p=0.07).Conclusions:Our results in Colombian patients indicate that GRB7 expression in HER2-positive breast tumors is not associated with unfavorable clinicopathological features.


Subject(s)
Female , Receptor, ErbB-2 , Ki-67 Antigen , GRB7 Adaptor Protein
5.
Mol Cancer ; 21(1): 8, 2022 01 03.
Article in English | MEDLINE | ID: mdl-34980129

ABSTRACT

BACKGROUND: Approximate 25% HER2-positive (HER2+) breast cancer (BC) patients treated with trastuzumab recurred rapidly. However, the mechanisms underlying trastuzumab resistance remained largely unclear. METHODS: Trastuzumab-resistant associated circRNAs were identified by circRNAs high-throughput screen and qRT-PCR in HER2+ breast cancer tissues with different trastuzumab response. The biological roles of trastuzumab-resistant associated circRNAs were detected by cell vitality assay, colony formation assay, Edu assay, patient-derived xenograft (PDX) models and orthotopic animal models. For mechanisms research, the co-immunoprecipitation, Western blot, immunofluorescence, and pull down assays confirmed the relevant mechanisms of circRNA and binding proteins. RESULTS: We identified a circRNA circCDYL2, which was overexpressed in trastuzumab-resistant patients, which conferred trastuzumab resistance in breast cancer cells in vitro and in vivo. Mechanically, circCDYL2 stabilized GRB7 by preventing its ubiquitination degradation and enhanced its interaction with FAK, which thus sustained the activities of downstream AKT and ERK1/2. Trastuzumab-resistance of HER2+ BC cells with high circCDYL2 could be reversed by FAK or GRB7 inhibitor. Clinically, HER2+ BC patients with high levels of circCDYL2 developed rapid recurrence and had shorter disease-free survival (DFS) and overall survival (OS) following anti-HER2 therapy compared to those with low circCDYL2. CONCLUSIONS: circCDYL2-GRB7-FAK complex plays a critical role in maintaining HER2 signaling, which contributes to trastuzumab resistance and circCDYL2 is a potential biomarker for trastuzumab-resistance in HER2+ BC patients.


Subject(s)
Breast Neoplasms/genetics , Co-Repressor Proteins/genetics , Drug Resistance, Neoplasm/genetics , Hydro-Lyases/genetics , RNA, Circular , Receptor, ErbB-2/metabolism , Signal Transduction , Animals , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Line, Tumor , Disease Management , Disease Susceptibility , Female , GRB7 Adaptor Protein/metabolism , Humans , Mice , Protein Binding , Proteolysis , Radiotherapy , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Ubiquitination
6.
Bioengineered ; 12(2): 12179-12190, 2021 12.
Article in English | MEDLINE | ID: mdl-34783299

ABSTRACT

Growth factor receptor bound protein 7 (GRB7) plays an important role in regulating the growth and metastasis of ovarian cancer. Angiogenesis is the basis for the growth, invasion, and metastasis of malignant tumors. In the current study, we aimed to determine whether GRB7 plays a role in regulating angiogenesis in ovarian cancer. Immunohistochemistry on tissue microarray showed that GRB7 and platelet endothelial cell adhesion molecule-1 (PECAM-1/CD31) protein expression were positively correlated in ovarian cancer tissues. GRB7 knockdown suppressed vascular endothelial growth factor A (VEGFA) expression and reduced VEGFA secretion. The effects of GRB7-silenced SKOV-3 cells on human umbilical vein endothelial cells (HUVECs) were evaluated using a transwell cell co-culture model, which showed that knockdown of GRB7 in SKOV-3 cells suppressed HUVEC proliferation, migration, invasion, and tube formation. Moreover, knockdown of GRB7 in SKOV-3 cells downregulated the expression of proteins associated with angiogenesis, including vascular endothelial growth factor receptor-2 (VEGFR2), mitogen-activated protein kinase kinase 1 (MAP2K1/MEK1), extracellular signal-regulated kinases 1 and 2 (ERK1/2), notch receptor 1 (NOTCH1), and delta-like canonical Notch ligand 4 (DLL4) in HUVECs. In conclusion, knockdown of GRB7 in ovarian cancer cells is an attractive potential therapeutic target for the suppression of angiogenesis in ovarian cancer. GRB7 may regulate angiogenesis through VEGFA/VEGFR2 signaling and its downstream pathways.


Subject(s)
GRB7 Adaptor Protein/metabolism , Gene Knockdown Techniques , Neovascularization, Pathologic/metabolism , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ovarian Neoplasms/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
8.
Int J Biol Sci ; 16(16): 3221-3230, 2020.
Article in English | MEDLINE | ID: mdl-33162827

ABSTRACT

Growth factor receptor-bound protein 7 (GRB7) has been found closely related to the occurrence and development of various tumors, but its function in bladder cancer has not yet been elucidated. The study is aiming at investigating the expression and function of GRB7 in bladder cancer. The Cancer Genome Atlas (TCGA) database was selected to analyze mRNA levels of GRB7 in bladder cancer. RT-qPCR and Western blot were conducted to detect the expression of GRB7 in normal bladder epithelial cells, seven bladder cancer cell lines and eight pairs of malignant/nonmalignant bladder tissues. The role of GRB7 in tumor proliferation and tumorigenesis was explored by establishing stable cells, in vitro cell experiments and in vivo xenograft models. The molecular regulation mechanism of GRB7 in bladder cancer was investigated by treatment with AKT inhibitor. GRB7 mRNA was upregulated in bladder cancer samples compared with that in normal tissue samples. Overexpressing GRB7 significantly promoted the proliferation and tumorigenesis of bladder cancer. However, silencing GRB7 played the retarding part. GRB7 promoted G1/S transition by activating the AKT pathway. Our results indicate that GRB7 plays an important role in promoting proliferation and tumorigenesis of bladder cancer.


Subject(s)
Carcinogenesis , Cell Proliferation , GRB7 Adaptor Protein/genetics , Proto-Oncogene Proteins c-akt/metabolism , Urinary Bladder Neoplasms/pathology , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred NOD , Mice, SCID , Signal Transduction , Urinary Bladder Neoplasms/genetics , Xenograft Model Antitumor Assays
9.
J Pathol ; 252(3): 317-329, 2020 11.
Article in English | MEDLINE | ID: mdl-32737994

ABSTRACT

Efficacious therapeutic approaches are urgently needed to improve outcomes in patients with oesophageal adenocarcinoma (OAC). However, oncogenic drivers amenable to targeted therapy are limited and their functional characterisation is essential. Among few targeted therapies available, anti-human epidermal growth factor receptor 2 (HER2) therapy showed only modest benefit for patients with OAC. Herein, we investigated the potential oncogenic role of growth factor receptor bound protein 7 (GRB7), which is reported to be co-amplified with HER2 (ERBB2) in OAC. GRB7 was highly expressed in 15% of OAC tumours, not all of which could be explained by co-amplification with HER2, and was associated with a trend for poorer overall survival. Knockdown of GRB7 decreased proliferation and clonogenic survival, and induced apoptosis. Reverse phase protein array (RPPA) analyses revealed a role for PI3K, mammalian target of rapamycin (mTOR), MAPK, and receptor tyrosine kinase signalling in the oncogenic action of GRB7. Furthermore, the GRB7 and HER2 high-expressing OAC cell line Eso26 showed reduced cell proliferation upon GRB7 knockdown but was insensitive to HER2 inhibition by trastuzumab. Consistent with this, GRB7 knockdown in vivo with an inducible shRNA significantly inhibited tumour growth in cell line xenografts. HER2 expression did not predict sensitivity to trastuzumab, with Eso26 xenografts remaining refractory to trastuzumab treatment. Taken together, our study provides strong evidence for an oncogenic role for GRB7 in OAC and suggests that targeting GRB7 may be a potential therapeutic strategy for this cancer. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Adenocarcinoma/metabolism , Biomarkers, Tumor/metabolism , Esophageal Neoplasms/metabolism , GRB7 Adaptor Protein/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Animals , Antineoplastic Agents, Immunological/therapeutic use , Blotting, Western , Cell Line, Tumor , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Prognosis , Receptor, ErbB-2/metabolism , Survival Analysis , Trastuzumab/therapeutic use
10.
Mol Cell Biochem ; 472(1-2): 209-218, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32577949

ABSTRACT

It is of great significance to explore the molecular mechanism of thyroid cancer (TC) pathogenesis for its improvement and therapy. Growth factor receptor bound protein-7 (GRB7) has been regarded as an important regulatory gene in the developments of various malignant tumors. Our study aimed to illustrate the role of GRB7 in the TC pathology mechanism. Firstly, GRB7 was found to be significantly upregulated in 49 cases of TC tissues and 5 TC cell lines by using real-time quantitative polymerase chain reaction (RT-qPCR) and western blotting. Silencing GRB7 with siRNA dramatically inhibited proliferation and induced cell cycle arrest in TC cells. Besides, GRB7 silence resulted in the decrease of adenosine triphosphate content, glucose uptake, and lactose production in TC cells and attenuated the activity and expression of mitochondrial respiratory complex. We also demonstrated that GRB7 downregulation increased the levels of Bax and caspase 3, and inhibited the expression of Bcl-2, suggesting the induced mitochondrial apoptosis. More importantly, our study proved that mitogen-activated protein kinase/extracellular-regulated protein kinases (MAPK/ERK) signaling played a crucial role in the regulation of GRB7 on TC cell functions. In general, the present research verified that GRB7 was upregulated during TC development and modulated the proliferation, cell cycle, and mitochondrial apoptosis of TC cells by activating MAPK/ERK pathway. This may provide a novel target for the therapeutic strategy of TC.


Subject(s)
Apoptosis , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , GRB7 Adaptor Protein/metabolism , Mitochondria/pathology , Mitogen-Activated Protein Kinases/metabolism , Thyroid Neoplasms/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Case-Control Studies , Cell Cycle , Extracellular Signal-Regulated MAP Kinases/genetics , GRB7 Adaptor Protein/genetics , Gene Expression Regulation, Neoplastic , Humans , Mitochondria/metabolism , Mitogen-Activated Protein Kinases/genetics , Prognosis , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Tumor Cells, Cultured
11.
Arch Biochem Biophys ; 687: 108386, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32360748

ABSTRACT

Growth factor receptor bound protein 7 (Grb7) is a mammalian adaptor protein participating in signaling pathways implicated in cell migration, metastatic invasion, cell proliferation and tumor-associated angiogenesis. We expressed tagged versions of wild type Grb7 and the mutant Grb7Δ, lacking its calmodulin-binding domain (CaM-BD), in human embryonic kidney (HEK) 293 cells and rat glioma C6 cells to identify novel binding partners using shot-gun proteomics. Among the new identified proteins, we validated the ubiquitin-ligase Nedd4 (neural precursor cell expressed developmentally down-regulated protein 4), the heat-shock protein Hsc70/HSPA8 (heat shock cognate protein 70) and the cell cycle regulatory protein caprin-1 (cytoplasmic activation/proliferation-associated protein 1) in rat glioma C6 cells. Our results suggest a role of Grb7 in pathways where these proteins are implicated. These include protein trafficking and degradation, stress-response, chaperone-mediated autophagy, apoptosis and cell proliferation.


Subject(s)
Cell Cycle Proteins/metabolism , GRB7 Adaptor Protein/metabolism , HSC70 Heat-Shock Proteins/metabolism , Nedd4 Ubiquitin Protein Ligases/metabolism , Animals , Calmodulin-Binding Proteins/genetics , Calmodulin-Binding Proteins/metabolism , Cell Line, Tumor , GRB7 Adaptor Protein/genetics , HEK293 Cells , Humans , Mutation , Protein Binding , Protein Domains/genetics , Protein Structure, Secondary , Proteomics , Rats
12.
Int J Mol Sci ; 21(4)2020 Feb 17.
Article in English | MEDLINE | ID: mdl-32079204

ABSTRACT

Grb7 is a signalling adapter protein that engages activated receptor tyrosine kinases at cellular membranes to effect downstream pathways of cell migration, proliferation and survival. Grb7's cellular location was shown to be regulated by the small calcium binding protein calmodulin (CaM). While evidence for a Grb7/CaM interaction is compelling, a direct interaction between CaM and purified Grb7 has not been demonstrated and quantitated. In this study we sought to determine this, and prepared pure full-length Grb7, as well as its RA-PH and SH2 subdomains, and tested for CaM binding using surface plasmon resonance. We report a direct interaction between full-length Grb7 and CaM that occurs in a calcium dependent manner. While no binding was observed to the SH2 domain alone, we observed a high micromolar affinity interaction between the Grb7 RA-PH domain and CaM, suggesting that the Grb7/CaM interaction is mediated through this region of Grb7. Together, our data support the model of a CaM interaction with Grb7 via its RA-PH domain.


Subject(s)
Calmodulin/genetics , GRB7 Adaptor Protein/genetics , Pleckstrin Homology Domains/genetics , Calmodulin/metabolism , Cell Movement , Cell Proliferation , Escherichia coli/genetics , Escherichia coli/metabolism , GRB7 Adaptor Protein/metabolism , Protein Binding , Protein Conformation , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Surface Plasmon Resonance , src Homology Domains/genetics
13.
Molecules ; 24(20)2019 Oct 17.
Article in English | MEDLINE | ID: mdl-31627265

ABSTRACT

Grb7 is an adapter protein, overexpressed in HER2+ve breast and other cancers, and identified as a therapeutic target. Grb7 promotes both proliferative and migratory cellular pathways through interaction of its SH2 domain with upstream binding partners including HER2, SHC, and FAK. Here we present the evaluation of a series of monocyclic and bicyclic peptide inhibitors that have been developed to specifically and potently target the Grb7 SH2-domain. All peptides tested were found to inhibit signaling in both ERK and AKT pathways in SKBR-3 and MDA-MB-231 cell lines. Proliferation, migration, and invasion assays revealed, however, that the second-generation bicyclic peptides were not more bioactive than the first generation G7-18NATE peptide, despite their higher in vitro affinity for the target. This was found not to be due to steric hindrance by the cell-permeability tag, as ascertained by ITC, but to differences in the ability of the bicyclic peptides to interact with and penetrate cellular membranes, as determined using SPR and mass spectrometry. These studies reveal that just small differences to amino acid composition can greatly impact the effectiveness of peptide inhibitors to their intracellular target and demonstrate that G7-18NATE remains the most effective peptide inhibitor of Grb7 developed to date.


Subject(s)
Antineoplastic Agents/pharmacology , Epithelial Cells/drug effects , GRB7 Adaptor Protein/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Peptides, Cyclic/pharmacology , Signal Transduction/drug effects , Amino Acid Sequence , Antineoplastic Agents/chemical synthesis , Binding Sites , Cell Line , Cell Line, Tumor , Cell Membrane Permeability , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , GRB7 Adaptor Protein/genetics , GRB7 Adaptor Protein/metabolism , Humans , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Peptides, Cyclic/chemical synthesis , Phosphatidylcholines/chemistry , Phosphatidylcholines/metabolism , Phosphatidylserines/chemistry , Phosphatidylserines/metabolism , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Structure-Activity Relationship , src Homology Domains/drug effects
14.
J Mol Recognit ; 32(11): e2803, 2019 11.
Article in English | MEDLINE | ID: mdl-31353673

ABSTRACT

It is well known the dimerization state of receptor tyrosine kinases (RTKs), in conjunction with binding partners such as the growth factor receptor bound protein 7 (Grb7) protein, plays an important role in cell signaling regulation. Previously, we proposed, downstream of RTKs, that the phosphorylation state of Grb7SH2 domain tyrosine residues could control Grb7 dimerization, and dimerization may be an important regulatory step in Grb7 binding to RTKs. In this manner, additional dimerization-dependent regulation could occur downstream of the membrane-bound kinase in RTK-mediated signaling pathways. Extrapolation to the full-length (FL) Grb7 protein, and the ability to test this hypothesis further, has been hampered by the availability of large quantities of pure and stable FL protein. Here, we report the biophysical characterization of the FL Grb7 protein and also a mutant representing a tyrosine-phosphorylated Grb7 protein form. Through size exclusion chromatography and analytical ultracentrifugation, we show the phosphorylated-tyrosine-mimic Y492E-FL-Grb7 protein (Y492E-FL-Grb7) is essentially monomeric at expected physiological concentrations. It has been shown previously the wild-type FL Grb7(WT-FLGrb7) protein is dimeric with a dissociation constant (Kd) of approximately 11µM. Our studies here measure a FL protein dimerization Kd of WT-FL-Grb7 within one order of magnitude at approximately 1µM. The approximate size and shape of the WT-FL-Grb7 in comparison the tyrosine-phosphorylation mimic Y492E-FL-Grb7 protein was determined by dynamic light scattering methods. In vitro phosphorylation of the Grb7SH2 domain indicates only one of the available tyrosine residues is phosphorylated, suggesting the same phosphorylation pattern could be relevant in the FL protein. The biophysical characterization studies in total are interpreted with a view towards understanding the functionally active Grb7 protein conformation.


Subject(s)
GRB7 Adaptor Protein/metabolism , Mutant Proteins/metabolism , Dynamic Light Scattering , GRB7 Adaptor Protein/chemistry , Humans , Molecular Weight , Mutant Proteins/chemistry , Phosphorylation , Phosphotyrosine/metabolism , Protein Domains , Protein Structure, Secondary
15.
J Cell Physiol ; 234(12): 23369-23378, 2019 12.
Article in English | MEDLINE | ID: mdl-31161615

ABSTRACT

The mechanisms of B-type Raf kinase (BRAF) V600E mutation in papillary thyroid cancer (PTC) remain to be elucidated. With the aim to investigate the key candidate genes distinctive to BRAFV600E -PTC, we analyzed the transcriptomics data from three microarray datasets (GSE27155, GSE54958, and GSE58545) and identified 491 differentially expressed genes (DEGs) between BRAFV600E -PTC and BRAFwild type -PTC. Functional enrichment analysis of DEGs revealed that negative regulation of wound healing may be involved in the BRAFV600E -related pathogenesis in PTC. Weighted gene coexpression network analysis revealed BRAFV600E -related coexpressed genes in PTC, from which hub genes were selected. The intersection of DEGs and hub genes revealed 31 candidates, wherein GRB7, SNAP25, SLC35F2, FAM155B, HGD, and ITPR1 were rendered the key candidate genes via receiver operating characteristic curve analysis. On further characterization, the six key genes displayed significantly different expression patterns at different cytomorphology, however, with no significant difference in overall survival. These results provide novel insights into the key genes distinctive to of BRAFV600E in PTC and might be suggestive as therapeutic targets for further application.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Profiling , Gene Regulatory Networks , Mutation , Proto-Oncogene Proteins B-raf/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/genetics , Transcriptome , GRB7 Adaptor Protein/genetics , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Homogentisate 1,2-Dioxygenase/genetics , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Membrane Transport Proteins/genetics , Oligonucleotide Array Sequence Analysis , Predictive Value of Tests , Signal Transduction , Synaptosomal-Associated Protein 25/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/pathology
16.
Cells ; 8(5)2019 05 10.
Article in English | MEDLINE | ID: mdl-31083325

ABSTRACT

The partner of activated epidermal growth factor receptor (EGFR), growth factor receptor bound protein-7 (Grb7), a functionally multidomain adaptor protein, has been demonstrated to be a pivotal regulator for varied physiological and pathological processes by interacting with phospho-tyrosine-related signaling molecules to affect the transmission through a number of signaling pathways. In particular, critical roles of Grb7 in erythroblastic leukemia viral oncogene homolog (ERBB) family-mediated cancer development and malignancy have been intensively evaluated. The overexpression of Grb7 or the coamplification/cooverexpression of Grb7 and members of the ERBB family play essential roles in advanced human cancers and are associated with decreased survival and recurrence of cancers, emphasizing Grb7's value as a prognostic marker and a therapeutic target. Peptide inhibitors of Grb7 are being tested in preclinical trials for their possible therapeutic effects. Here, we review the molecular, functional, and clinical aspects of Grb7 in ERBB family-mediated cancer development and malignancy with the aim to reveal alternative and effective therapeutic strategies.


Subject(s)
Biomarkers, Tumor , GRB7 Adaptor Protein , Neoplasms/metabolism , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/physiology , ErbB Receptors/metabolism , GRB7 Adaptor Protein/chemistry , GRB7 Adaptor Protein/metabolism , GRB7 Adaptor Protein/physiology , Humans , Neoplasms/therapy , Signal Transduction
17.
Int J Mol Med ; 43(6): 2352-2360, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31017268

ABSTRACT

Emerging evidence has demonstrated that microRNAs (miRNAs/miRs) have various biological functions in the development of human epidermal growth factor receptor 2 (HER2) positive breast cancer. The aim of the present study is to reveal the mechanism of miR­193a­3p inhibiting the progress of HER2 positive breast cancer. The expression of miR­193a­3p was evaluated by quantitative polymerase chain reaction (PCR). The methylation status of miR­193a­3p was evaluated by PCR and pyrosequencing analysis. Overexpression of miR­193a­3p and growth factor receptor bound protein 7 (GRB7) combined with in vitro tumorigenic assays were conducted to determine the carcinostatic capacities of miR­193a­3p in HER2 positive breast cancer cells. The association between miR­193a­3p and GRB7 was determined by luciferase reporter assay. Protein level was evaluated using western blot analysis. miR­193a­3p was downregulated in HER2 positive breast cancer cells and clinical tissues. Methylation­mediated silencing led to decreased expression of miR­193a­3p in HER2 positive breast cancer. Overexpression of miR­193a­3p could inhibit proliferation, migration and invasion of breast cancer cells. Overexpression of GRB7 could abolish this effect. miR­193a­3p could directly target the 3' untranslated region of GRB7. miR­193a­3p could directly or indirectly target extracellular signal­regulated kinase 1/2 (ERK1/2) and forkhead box M1 (FOXM1) signaling. In conclusion, it was identified that silencing of miR­193a­3p through hypermethylation can promote HER2 positive breast cancer progress by targeting GRB7, ERK1/2 and FOXM1 signaling. The function of miR­193a­3p in HER2 positive breast cancer implicates its potential application in therapy.


Subject(s)
Breast Neoplasms/genetics , GRB7 Adaptor Protein/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neoplasm Invasiveness/genetics , Receptor, ErbB-2/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Epigenesis, Genetic , Female , Humans , Neoplasm Invasiveness/pathology
18.
Mol Carcinog ; 58(5): 699-707, 2019 05.
Article in English | MEDLINE | ID: mdl-30604896

ABSTRACT

GRB7 gene encodes a multi-domain signal transduction molecule and is part of the core of the HER-2 amplicon. GRB7 is commonly co-amplified and overexpressed with HER-2 in human breast cancer. This study addresses the role of GRB7 in HER-2 positive human breast cancers resistant to HER-2 targeted therapy. HCC1954, 21MT1, and JIMT1 are basal like HER-2 positive breast cancer cell lines based on expression profiling. These three cell lines are resistant to trastuzumab and lapatinib treatment. Knockdown of GRB7 protein expression with siRNA transfection as well as lentiviral vector mediated shRNA over-expression decreased the growth of HCC1954, 21MT1, and JIMT1 cells in vitro and the growth of tumor xenografts these cells formed in animal models. When assayed by ki-67 staining and TUNEL assay, the mechanism of reduced tumor xenograft growth appeared to be distinct. Reduced proliferation and increased apoptosis were seen in 21MT1 cells, while reduced proliferation was seen in HCC1954 cells and increased apoptosis in JIMT1 cells. Phospho-proteome profiling found HER-1 tyrosine phosphorylation was reduced with GRB7 knock down in JIMT1 cells. Immuno-blotting and immuno-precipitation experiments found HER-1 phosphorylation was reduced with GRB7 knock down in all three cell lines. HER-1 knock down via siRNA transient transfection as well as blocking HER-1 function with panitumumab decreased proliferation of all three cell lines in vitro. Our study finds that GRB7 has an essential growth promoting function which is mediated in part by HER-1 activation. The potential of HER-1 targeting in therapy resistant HER-2 positive breast cancer merits further study.


Subject(s)
Breast Neoplasms/pathology , Cell Proliferation , GRB7 Adaptor Protein/metabolism , Neoplasms, Basal Cell/pathology , Receptor, ErbB-2/metabolism , Animals , Apoptosis , Breast Neoplasms/metabolism , Cell Movement , ErbB Receptors/metabolism , Female , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Basal Cell/metabolism , Phosphorylation , Signal Transduction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
19.
Theranostics ; 8(2): 423-436, 2018.
Article in English | MEDLINE | ID: mdl-29290818

ABSTRACT

Human growth factor receptor-bound protein-7 (GRB7) is a pivotal mediator involved in receptor tyrosine kinase signaling and governing diverse cellular processes. Aberrant upregulation of GRB7 is frequently associated with the progression of human cancers. However, the molecular mechanisms leading to the upregulation of GRB7 remain largely unknown. Here, we propose that the epigenetic modification of GRB7 at the post-transcriptional level may be a crucial factor leading to GRB7 upregulation in ovarian cancers. Methods: The upstream miRNA regulators were predicted by in silico analysis. Expression of GRB7 was examined by qPCR, immunoblotting and immunohistochemical analyses, while miR-193a-3p levels were evaluated by qPCR and in situ hybridization in ovarian cancer cell lines and clinical tissue arrays. MS-PCR and pyrosequencing analyses were used to assess the methylation status of miR-193a-3p. Stable overexpression or gene knockdown and Tet-on inducible approaches, in combination with in vitro and in vivo tumorigenic assays, were employed to investigate the functions of GRB7 and miR-193a-3p in ovarian cancer cells. Results: Both miR-193a-3p and its isoform, miR-193b-3p, directly targeted the 3' UTR of GRB7. However, only miR-193a-3p showed a significantly inverse correlation with GRB7-upregulated ovarian cancers. Epigenetic studies revealed that methylation-mediated silencing of miR-193a-3p led to a stepwise decrease in miR-193a-3p expression from low to high-grade ovarian cancers. Intriguingly, miR-193a-3p not only modulated GRB7 but also ERBB4, SOS2 and KRAS in the MAPK/ERK signaling pathway to enhance the oncogenic properties of ovarian cancer cells in vitro and in vivo. Conclusion: These findings suggest that epigenetic silencing of miR-193a-3p by DNA hypermethylation is a dynamic process in ovarian cancer progression, and miR-193a-3p may be explored as a promising miRNA replacement therapy in this disease.


Subject(s)
Cell Movement/genetics , GRB7 Adaptor Protein/genetics , Gene Silencing/physiology , MAP Kinase Signaling System/genetics , MicroRNAs/genetics , Ovarian Neoplasms/genetics , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Methylation , Mice , Mice, Inbred BALB C , Mice, Nude , RNA Interference/physiology , Signal Transduction/genetics , Up-Regulation/genetics
20.
J Med Chem ; 60(22): 9349-9359, 2017 11 22.
Article in English | MEDLINE | ID: mdl-29083893

ABSTRACT

Grb7 is a signaling protein with critical roles in tumor cell proliferation and migration and an established cancer therapeutic target. Here we explore chemical space to develop a new bicyclic peptide inhibitor, incorporating thioether and lactam linkers that binds with affinity (KD = 1.1 µM) and specificity to the Grb7-SH2 domain. Structural analysis of the Grb7-SH2/peptide complex revealed an unexpected binding orientation underlying the binding selectivity by this new scaffold. We further incorporated carboxymethylphenylalanine and carboxyphenylalanine phosphotyrosine mimetics and arrived at an optimized inhibitor that potently binds Grb7-SH2 (KD = 0.13 µM) under physiological conditions. X-ray crystal structures of these Grb7-SH2/peptide complexes reveal the structural basis for the most potent and specific inhibitors of Grb7 developed to date. Finally, we demonstrate that cell permeable versions of these peptides successfully block Grb7 mediated interactions in a breast cancer cell line, establishing the potential of these peptides in the development of novel therapeutics targeted to Grb7.


Subject(s)
GRB7 Adaptor Protein/antagonists & inhibitors , Peptides, Cyclic/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Drug Discovery , Focal Adhesion Kinase 1/metabolism , GRB7 Adaptor Protein/chemistry , GRB7 Adaptor Protein/metabolism , Humans , Lactams/chemical synthesis , Lactams/chemistry , Lactams/pharmacology , Ligands , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/chemistry , Phosphates/chemistry , Protein Conformation , Receptor, ErbB-2/metabolism , Shc Signaling Adaptor Proteins/metabolism , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...