Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
BMC Biol ; 22(1): 127, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816743

ABSTRACT

BACKGROUND: Optimal size at birth dictates perinatal survival and long-term risk of developing common disorders such as obesity, type 2 diabetes and cardiovascular disease. The imprinted Grb10 gene encodes a signalling adaptor protein capable of inhibiting receptor tyrosine kinases, including the insulin receptor (Insr) and insulin-like growth factor type 1 receptor (Igf1r). Grb10 restricts fetal growth such that Grb10 knockout (KO) mice are at birth some 25-35% larger than wild type. Using a mouse genetic approach, we test the widely held assumption that Grb10 influences growth through interaction with Igf1r, which has a highly conserved growth promoting role. RESULTS: Should Grb10 interact with Igf1r to regulate growth Grb10:Igf1r double mutant mice should be indistinguishable from Igf1r KO single mutants, which are around half normal size at birth. Instead, Grb10:Igf1r double mutants were intermediate in size between Grb10 KO and Igf1r KO single mutants, indicating additive effects of the two signalling proteins having opposite actions in separate pathways. Some organs examined followed a similar pattern, though Grb10 KO neonates exhibited sparing of the brain and kidneys, whereas the influence of Igf1r extended to all organs. An interaction between Grb10 and Insr was similarly investigated. While there was no general evidence for a major interaction for fetal growth regulation, the liver was an exception. The liver in Grb10 KO mutants was disproportionately overgrown with evidence of excess lipid storage in hepatocytes, whereas Grb10:Insr double mutants were indistinguishable from Insr single mutants or wild types. CONCLUSIONS: Grb10 acts largely independently of Igf1r or Insr to control fetal growth and has a more variable influence on individual organs. Only the disproportionate overgrowth and excess lipid storage seen in the Grb10 KO neonatal liver can be explained through an interaction between Grb10 and the Insr. Our findings are important for understanding how positive and negative influences on fetal growth dictate size and tissue proportions at birth.


Subject(s)
Fetal Development , GRB10 Adaptor Protein , Mice, Knockout , Receptor, IGF Type 1 , Receptor, Insulin , Animals , GRB10 Adaptor Protein/genetics , GRB10 Adaptor Protein/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Mice , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Fetal Development/genetics , Genomic Imprinting , Female , Male , Insulin-Like Peptides
2.
Mol Med ; 29(1): 133, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37789274

ABSTRACT

BACKGROUND: Rab-interacting lysosomal protein (RILP) contains an alpha-helical coil with an unexplored biological function in osteosarcoma. This study investigated the expression of RILP in osteosarcoma cells and tissues to determine the effect of RILP on the biological behaviors of osteosarcoma cells and the underlying mechanism. METHODS: Tumor Immune Estimation Resource (TIMER) database, The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) database were used for bioinformatic analysis. Co-immunoprecipitation experiment was used to determine whether the two proteins were interacting. In functional tests, cell counting kit-8 (CCK-8) assay, colony formation assay, wound healing assay, transwell invasion assay, Immunofluorescence (IF) assay and immunohistochemical (IHC) assay were performed. RESULTS: Overexpression of RILP significantly inhibited proliferation and impaired metastasis ability of osteosarcoma cells, while silencing of RILP showed the opposite trend. RNA-seq data analysis was applied in 143B cells and pathway enrichment analysis revealed that differentially expressed genes were mainly enriched in the PI3K/AKT pathway. We further verified that overexpression of RILP restrained the PI3K/AKT/mTOR signaling pathway and induced autophagy in osteosarcoma cells, while the opposite trend was observed when PI3K pathway activator 740Y-P was used. 3-Methyladenine (3-MA), a selective autophagy inhibitor, partially attenuated the inhibitory effect of RILP on the migration and invasion ability of osteosarcoma cells, suggesting the involvement of autophagy in epithelial-mesenchymal transition regulation in osteosarcoma cells. Growth factor receptor binding protein-10 (Grb10), an adaptor protein, was confirmed as a potential target of RILP to restrain the PI3K/AKT signaling pathway. We subcutaneously injected stably overexpressing 143B osteosarcoma cells into nude mice and observed that overexpression of RILP inhibited tumor growth by inhibiting the PI3K/AKT/mTOR pathway. CONCLUSION: Our study revealed that the expression of RILP was associated with favorable prognosis of osteosarcoma and RILP inhibits proliferation, migration, and invasion and promotes autophagy in osteosarcoma cells via Grb10-mediated inhibition of the PI3K/AKT/mTOR signaling pathway. In the future, targeting RILP may be a potential strategy for osteosarcoma treatment.


Subject(s)
Bone Neoplasms , Osteosarcoma , Animals , Mice , Apoptosis , Bone Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , GRB10 Adaptor Protein/metabolism , GRB10 Adaptor Protein/pharmacology , Mice, Nude , Osteosarcoma/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Humans
3.
Nat Metab ; 5(1): 147-164, 2023 01.
Article in English | MEDLINE | ID: mdl-36593271

ABSTRACT

Leptin acts on hypothalamic neurons expressing agouti-related protein (AgRP) or pro-opiomelanocortin (POMC) to suppress appetite and increase energy expenditure, but the intracellular mechanisms that modulate central leptin signalling are not fully understood. Here we show that growth factor receptor-bound protein 10 (Grb10), an adaptor protein that binds to the insulin receptor and negatively regulates its signalling pathway, can interact with the leptin receptor and enhance leptin signalling. Ablation of Grb10 in AgRP neurons promotes weight gain, while overexpression of Grb10 in AgRP neurons reduces body weight in male and female mice. In parallel, deletion or overexpression of Grb10 in POMC neurons exacerbates or attenuates diet-induced obesity, respectively. Consistent with its role in leptin signalling, Grb10 in AgRP and POMC neurons enhances the anorexic and weight-reducing actions of leptin. Grb10 also exaggerates the inhibitory effects of leptin on AgRP neurons via ATP-sensitive potassium channel-mediated currents while facilitating the excitatory drive of leptin on POMC neurons through transient receptor potential channels. Our study identifies Grb10 as a potent leptin sensitizer that contributes to the maintenance of energy homeostasis by enhancing the response of AgRP and POMC neurons to leptin.


Subject(s)
Leptin , Pro-Opiomelanocortin , Mice , Male , Female , Animals , Agouti-Related Protein/metabolism , Leptin/metabolism , Pro-Opiomelanocortin/metabolism , GRB10 Adaptor Protein/metabolism , Weight Loss
4.
Oxid Med Cell Longev ; 2022: 3522492, 2022.
Article in English | MEDLINE | ID: mdl-35571247

ABSTRACT

Circular RNAs (circRNAs) are abundant in the brain and contribute to central nervous system diseases; however, the exact roles of circRNAs in human traumatic brain injury (TBI) have not been established. In this study, we used a competing endogenous RNA (ceRNA) chipset as well as in vitro and in vivo assays to characterize differentially expressed circRNAs in TBI. We detected 3035 differentially expressed circRNAs in the severe TBI group, 2362 in the moderate group, and 433 in the mild group. A ceRNA network was constructed. The circRNA has_circ_0020269 (circHtra1) was significantly upregulated after brain insults and was correlated with the severity of injury. circHtra1 inhibited cell proliferation and promoted apoptosis, and its knockdown reversed these effects. Further analyses revealed that circHtra1 functions as a miR-3960 sponge and increases the expression of GRB10, which is involved in NK cell infiltration after TBI. circHtra1 was identified as a target of the IGF-1/ADAR1 axis. Reduced expression of ADAR1 (involved in A-to-I editing) after brain insults upregulated circHtra1. Our results show that circHtra1 promotes neuronal loss by sponging miR-3960 and regulating GRB10 and apoptosis during brain insults. In addition, A-to-I editing could regulate circRNA expression profiles after TBI, and circHtra1 is a potential therapeutic target.


Subject(s)
Brain Injuries, Traumatic , MicroRNAs , Apoptosis/genetics , Brain Injuries, Traumatic/genetics , Brain Injuries, Traumatic/metabolism , Cell Proliferation/genetics , GRB10 Adaptor Protein/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics
5.
Theranostics ; 12(6): 2613-2630, 2022.
Article in English | MEDLINE | ID: mdl-35401828

ABSTRACT

Background: Insulin-like growth factor 1 receptor (IGF-1R) expression and signaling play important roles in promotion of skin cancer progression. Identification of signaling pathways that regulate IGF-1R is crucial for understanding the pathogenesis and therapeutic treatment of skin cancer. Methods: Molecular, cellular and genetic approaches were used to investigate the function of PINCH-1 in regulation of IGF-1R expression and skin cell behavior. Furthermore, conditional PINCH-1 knockout mouse and carcinogen (7, 12-dimethylbenz[a]anthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA))-induced skin cancer model were employed to determine the function of PINCH-1 in regulation of IGF-1R expression and skin carcinogenesis in vivo. Results: Knockdown of PINCH-1 from HaCaT keratinocytes or A431 squamous carcinoma cells diminished IGF-1R levels, suppressed cell proliferation and increased apoptosis. Re-expression of PINCH-1 in PINCH-1 knockdown cells restored IGF-1R expression, cell proliferation and survival. Furthermore, depletion of NEDD4 effectively reversed PINCH-1 deficiency-induced down-regulation of IGF-1R expression, cell proliferation and survival. Conditional knockout of PINCH-1 from keratin 5 (K5) positive keratinocytes in mice, like depletion of PINCH-1 from keratinocytes in culture, reduced the IGF-1R level. Using a mouse model of DMBA/TPA-induced skin cancer, we show that the levels of both PINCH-1 and IGF-1R were significantly increased in response to treatment with the carcinogens. Genetic ablation of PINCH-1 from the epidermis markedly reduced the IGF-1R expression and cell proliferation despite stimulation with DMBA/TPA, resulting in resistance to chemical carcinogen-induced skin cancer initiation and progression. Conclusions: Our results reveal a PINCH-1-NEDD4-IGF-1R signaling axis that is critical for promotion of skin tumorigenesis and suggest a new strategy for therapeutic control of skin cancer progression.


Subject(s)
Receptor, IGF Type 1 , Skin Neoplasms , Animals , Carcinogenesis/pathology , Carcinogens/metabolism , Cell Proliferation , GRB10 Adaptor Protein/metabolism , GRB10 Adaptor Protein/pharmacology , Keratinocytes , Mice , Receptor, IGF Type 1/genetics , Skin/pathology , Skin Neoplasms/chemically induced , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Tetradecanoylphorbol Acetate/adverse effects , Tetradecanoylphorbol Acetate/metabolism
6.
Brain Res Bull ; 181: 12-20, 2022 04.
Article in English | MEDLINE | ID: mdl-35065184

ABSTRACT

Type 2 diabetes is a major factor contributing to cognitive decline and Alzheimer's disease (AD). Treadmill running is considered to be a critical approach for mice and rats to lower blood sugar and improve learning and memory capacity. The growth factor receptor-bound protein 10 (Grb10) has been proposed to inhibit insulin signaling and defective brain insulin signaling resulted in the cognitive deficits in patients with AD. However, the positive roles of treadmill training on diabetic- related impaired cognitive function and their molecular mechanisms remain unclear. Here, to investigate whether there was neuroprotective effects of treadmill training on impaired cognitive function caused by diabetes, the rats were injected intraperitoneally with streptozotocin at a dose of 30 mg/kg to establish diabetic model (DM). We found that higher Grb10, BACE1 and PHF10 protein levels in the hippocampus of DM rats, lower phosphorylation IGF-1Rß and IRS-1(ser307). However, 8 weeks treadmill training effectively reduced abnormal Grb10, enhanced postsynaptic density protein PSD-93, PSD-95, SYN expressions of hippocampus, restored PI3K/Akt/ERK and mTOR/AMPK signaling, thus alleviated spatial learning and memory deficit, compared with DM group. Additionally, treadmill training also increased GLUT4 transportation. Overall, our findings suggest that treadmill intervention improved cognitive impairments caused by diabetes disease partly through modulating Grb10/ PI3K/Akt/ERK as well as mTOR/AMPK signaling.


Subject(s)
Cognitive Dysfunction/therapy , Diabetes Complications/therapy , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Type 2/complications , Exercise Therapy , GRB10 Adaptor Protein/metabolism , Glucose Transporter Type 4/drug effects , Physical Conditioning, Animal , Running , Animals , Antibiotics, Antineoplastic/administration & dosage , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/metabolism , Diabetes Complications/chemically induced , Diabetes Complications/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/metabolism , Physical Conditioning, Animal/physiology , Rats , Running/physiology , Streptozocin/administration & dosage
7.
J Genet Genomics ; 49(3): 208-216, 2022 03.
Article in English | MEDLINE | ID: mdl-34861413

ABSTRACT

Decreased functional ß-cell mass is the hallmark of diabetes, but the cause of this metabolic defect remains elusive. Here, we show that the levels of the growth factor receptor-bound protein 10 (GRB10), a negative regulator of insulin and mTORC1 signaling, are markedly induced in islets of diabetic mice and high glucose-treated insulinoma cell line INS-1 cells. ß-cell-specific knockout of Grb10 in mice increased ß-cell mass and improved ß-cell function. Grb10-deficient ß-cells exhibit enhanced mTORC1 signaling and reduced ß-cell dedifferentiation, which could be blocked by rapamycin. On the contrary, Grb10 overexpression induced ß-cell dedifferentiation in MIN6 cells. Our study identifies GRB10 as a critical regulator of ß-cell dedifferentiation and ß-cell mass, which exerts its effect by inhibiting mTORC1 signaling.


Subject(s)
Diabetes Mellitus, Experimental , GRB10 Adaptor Protein , Animals , Cell Dedifferentiation/genetics , Cell Proliferation/genetics , GRB10 Adaptor Protein/genetics , GRB10 Adaptor Protein/metabolism , Insulin/metabolism , Mice
8.
Hematol Oncol ; 39(3): 390-400, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33969901

ABSTRACT

The exosomes are involved in intercellular communication via RNA trafficking in human diseases. Hsa_circ_0009910 (circ_0009910) is a novel leukemia-related circular RNA. However, the mechanism of circ_0009910 in acute myeloid leukemia (AML) cell-to-cell communication remained obscure. Expression of circ_0009910, miRNA (miR)-5195-3p and growth factor receptor-bound protein 10 (GRB10) was detected by quantitative real-time polymerase chain reaction and Western blotting. A stable cell coculture model was established and functional experiment was performed using Cell Counting Kit-8 assay, flow cytometry, and Western blotting. The interaction among circ_0009910, miR-5195-3p and GRB10 was confirmed by dual-luciferase reporter assay and RNA immunoprecipitation. As a result, circ_0009910 was upregulated in AML bone marrows and cells (HL-60 and MOLM-13), even higher in AML cells-derived exosomes. Functionally, blocking circ_0009910 via small interfering RNA (siRNA) suppressed cell proliferation and cell cycle progression, but facilitated apoptosis rate of HL-60 and MOLM-13 cells, accompanied with lower B-cell lymphoma 2 (Bcl-2) level and higher Bcl-2-associated X protein (Bax) level. circ_0009910 shuttled via exosomes negatively regulated miR-5195-3p expression by target binding. Furthermore, circ_0009910 knockdown via exosomes and miR-5195-3p overexpression via mimic resulted in similar results of circ_0009910 siRNA in proliferation, apoptosis and cell cycle progression of AML cells. Meanwhile, the role of circ_0009910 knockdown in AML cells was partially reversed by miR-5195-3p deletion, and restoring GRB10 could abrogate miR-5195-3p effect as well. Notably, GRB10 was a downstream target of miR-5195-3p. circ_0009910-containing exosomes mediated proliferation, apoptosis and cell cycle progression of AML cells partially through miR-5195-3p/GRB10 axis.


Subject(s)
Apoptosis , Cell Cycle , Exosomes/metabolism , GRB10 Adaptor Protein/metabolism , Leukemia, Myeloid, Acute/metabolism , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , RNA, Circular/metabolism , RNA, Neoplasm/metabolism , Signal Transduction , Adult , Female , Humans , Male , Middle Aged
9.
Int J Cancer ; 148(2): 469-480, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33038264

ABSTRACT

Prostate cancer (PCa) progression is driven by androgen receptor (AR) signaling. Unfortunately, androgen-deprivation therapy and the use of even more potent AR pathway inhibitors (ARPIs) cannot bring about a cure. ARPI resistance (ie, castration-resistant PCa, CRPC) will inevitably develop. Previously, we demonstrated that GRB10 is an AR transcriptionally repressed gene that functionally contributes to CRPC development and ARPI resistance. GRB10 expression is elevated prior to CRPC development in our patient-derived xenograft models and is significantly upregulated in clinical CRPC samples. Here, we analyzed transcriptomic data from GRB10 knockdown in PCa cells and found that AR signaling is downregulated. While the mRNA expression of AR target genes decreased upon GRB10 knockdown, AR expression was not affected at the mRNA or protein level. We further found that phosphorylation of AR serine 81 (S81), which is critical for AR transcriptional activity, is decreased by GRB10 knockdown and increased by its overexpression. Luciferase assay using GRB10-knockdown cells also indicate reduced AR activity. Immunoprecipitation coupled with mass spectrometry revealed an interaction between GRB10 and the PP2A complex, which is a known phosphatase of AR. Further validations and analyses showed that GRB10 binds to the PP2Ac catalytic subunit with its PH domain. Mechanistically, GRB10 knockdown increased PP2Ac protein stability, which in turn decreased AR S81 phosphorylation and reduced AR activity. Our findings indicate a reciprocal feedback between GRB10 and AR signaling, implying the importance of GRB10 in PCa progression.


Subject(s)
GRB10 Adaptor Protein/metabolism , Prostatic Neoplasms/metabolism , Protein Phosphatase 2/metabolism , Receptors, Androgen/metabolism , Animals , Cell Line, Tumor , GRB10 Adaptor Protein/genetics , Gene Knockdown Techniques , HEK293 Cells , Heterografts , Humans , Male , Mice , Prostatic Neoplasms/genetics , Protein Phosphatase 2/antagonists & inhibitors , Signal Transduction
10.
Commun Biol ; 3(1): 157, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32242137

ABSTRACT

Aging induces cellular and molecular changes including modification of stem cell pools. In particular, alterations in aging neural stem cells (NSCs) are linked to age-related cognitive decline which can be modulated by lifestyle. Nutrient-sensing pathways provide a molecular basis for the link between lifestyle and cognitive decline. Adopting a back-translation strategy using stem cell biology to inform epidemiological analyses, here we show associations between cellular readouts of NSC maintenance and expression levels of nutrient-sensing genes following NSC exposure to aging human serum as well as morphological and gene expression alterations following repeated passaging. Epidemiological analyses on the identified genes showed associations between polymorphisms in SIRT1 and ABTB1 and cognitive performance as well as interactions between SIRT1 genotype and physical activity and between GRB10 genotype and adherence to a Mediterranean diet. Our study contributes to the understanding of neural stem cell molecular mechanisms underlying human cognitive aging and hints at lifestyle modifiable factors.


Subject(s)
Cellular Senescence , Cognitive Aging , Diet, Healthy , Exercise , Healthy Aging , Hippocampus/metabolism , Neural Stem Cells/metabolism , Adult , Age Factors , Aged , Aged, 80 and over , Cell Line , Cellular Senescence/genetics , Diet Surveys , Diet, Mediterranean , Energy Intake , Female , GRB10 Adaptor Protein/genetics , GRB10 Adaptor Protein/metabolism , Gene Expression Regulation , Gene-Environment Interaction , Genetic Association Studies , Healthy Aging/genetics , Healthy Aging/metabolism , Healthy Aging/pathology , Hippocampus/pathology , Humans , Male , Middle Aged , Neural Stem Cells/pathology , Polymorphism, Single Nucleotide , Repressor Proteins/genetics , Repressor Proteins/metabolism , Risk Reduction Behavior , Sirtuin 1/genetics , Sirtuin 1/metabolism , United Kingdom , Young Adult
11.
J Diabetes Res ; 2019: 9582714, 2019.
Article in English | MEDLINE | ID: mdl-31179345

ABSTRACT

Exposure to low concentration of the common food additive carrageenan (10 mg/L) for only six days led to glucose intolerance and insulin resistance in the C57BL/6J mouse. Longer exposure produced fasting hyperglycemia but with no increase in weight, in contrast to the HFD. Glucose intolerance was attributable to carrageenan-induced inflammation and to increased expression of GRB10. Both HFD and carrageenan increased p(Ser32)-IκBα and p(Ser307)-IRS1, and the increases were greater following the combined exposure. The effects of carrageenan were inhibited by the combination of the free radical inhibitor Tempol and BCL10 siRNA, which had no impact on the HFD-mediated increase. In contrast, the PKC inhibitor sotrastaurin blocked the HFD-induced increases, without an effect on the carrageenan-mediated effects. HFD had no impact on the expression of GRB10. Both carrageenan and high fat increased hepatic infiltration by F4/80-positive macrophages. Serum galectin-3 and galectin-3 binding to the insulin receptor increased by carrageenan and by HFD. Tyrosine phosphorylation of the insulin receptor declined following either exposure and was further reduced by their combination. Carrageenan reduced the activity of the enzyme N-acetylgalactosamine-4-sulfatase (ARSB; arylsulfatase B), which was unchanged following HFD. Dietary exposure to both high fat and carrageenan can impair insulin signaling through both similar and distinct mechanisms.


Subject(s)
Carrageenan/pharmacology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/therapy , Diet, High-Fat/adverse effects , Insulin Resistance , Animals , B-Cell CLL-Lymphoma 10 Protein/metabolism , Cyclic N-Oxides/pharmacology , Disease Models, Animal , GRB10 Adaptor Protein/metabolism , Galectin 3/metabolism , Gene Expression Regulation , Glucose Intolerance , Hep G2 Cells , Humans , Inflammation , Insulin/metabolism , Liver/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , N-Acetylgalactosamine-4-Sulfatase/metabolism , Protein Binding , RNA, Small Interfering/metabolism , Signal Transduction , Spin Labels , Triglycerides/metabolism
12.
Elife ; 82019 04 29.
Article in English | MEDLINE | ID: mdl-31033440

ABSTRACT

Iron and heme play central roles in the production of red blood cells, but the underlying mechanisms remain incompletely understood. Heme-regulated eIF2α kinase (HRI) controls translation by phosphorylating eIF2α. Here, we investigate the global impact of iron, heme, and HRI on protein translation in vivo in murine primary erythroblasts using ribosome profiling. We validate the known role of HRI-mediated translational stimulation of integratedstressresponse mRNAs during iron deficiency in vivo. Moreover, we find that the translation of mRNAs encoding cytosolic and mitochondrial ribosomal proteins is substantially repressed by HRI during iron deficiency, causing a decrease in cytosolic and mitochondrial protein synthesis. The absence of HRI during iron deficiency elicits a prominent cytoplasmic unfolded protein response and impairs mitochondrial respiration. Importantly, ATF4 target genes are activated during iron deficiency to maintain mitochondrial function and to enable erythroid differentiation. We further identify GRB10 as a previously unappreciated regulator of terminal erythropoiesis.


Subject(s)
Erythropoiesis/physiology , Heme/metabolism , Iron/metabolism , Mitochondria/metabolism , Proteostasis/physiology , eIF-2 Kinase/metabolism , Activating Transcription Factor 4/genetics , Anemia, Iron-Deficiency , Animals , Cell Differentiation , Erythroblasts , Eukaryotic Initiation Factor-2/metabolism , GRB10 Adaptor Protein/genetics , GRB10 Adaptor Protein/metabolism , Mice , Mice, Knockout , Oxygen/metabolism , Phosphorylation , Protein Biosynthesis , Ribosomal Proteins , Unfolded Protein Response , eIF-2 Kinase/genetics
13.
FASEB J ; 33(3): 3198-3211, 2019 03.
Article in English | MEDLINE | ID: mdl-30379590

ABSTRACT

Growth factor receptor-binding protein 10 (GRB10) is a well-known adaptor protein and a recently identified substrate of the mammalian target of rapamycin (mTOR). Depletion of GRB10 increases insulin sensitivity and overexpression suppresses PI3K/Akt signaling. Because the major reason for the limited efficacy of PI3K/Akt-targeted therapies in prostate cancer (PCa) is loss of mTOR-regulated feedback suppression, it is therefore important to assess the functional importance and regulation of GRB10 under these conditions. On the basis of these background observations, we explored the status and functional impact of GRB10 in PCa and found maximum expression in phosphatase and tensin homolog (PTEN)-deficient PCa. In human PCa samples, GRB10 inversely correlated with PTEN and positively correlated with pAKT levels. Knockdown of GRB10 in nontumorigenic PTEN null mouse embryonic fibroblasts and tumorigenic PCa cell lines reduced Akt phosphorylation and selectively activated a panel of receptor tyrosine kinases. Similarly, overexpression of GRB10 in PTEN wild-type PCa cell lines accelerated tumorigenesis and induced Akt phosphorylation. In PTEN wild-type PCa, GRB10 overexpression promoted mediated PTEN interaction and degradation. PI3K (but not mTOR) inhibitors reduced GRB10 expression, suggesting primarily PI3K-driven regulation of GRB10. In summary, our results suggest that GRB10 acts as a major downstream effector of PI3K and has tumor-promoting effects in prostate cancer.-Khan, M. I., Al Johani, A., Hamid, A., Ateeq, B., Manzar, N., Adhami, V. M., Lall, R. K., Rath, S., Sechi, M., Siddiqui, I. A., Choudhry, H., Zamzami, M. A., Havighurst, T. C., Huang, W., Ntambi, J. M., Mukhtar, H. Proproliferatve function of adaptor protein GRB10 in prostate carcinoma.


Subject(s)
GRB10 Adaptor Protein/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Carcinogens/antagonists & inhibitors , Carcinogens/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cell Proliferation/physiology , Fibroblasts/cytology , Fibroblasts/metabolism , GRB10 Adaptor Protein/antagonists & inhibitors , GRB10 Adaptor Protein/genetics , Gene Knockdown Techniques , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Models, Biological , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/genetics , RNA, Messenger , Signal Transduction
14.
Arch Gynecol Obstet ; 298(6): 1219-1227, 2018 12.
Article in English | MEDLINE | ID: mdl-30251157

ABSTRACT

PURPOSE: Grb10 is a key imprinted gene that is suspected to have a role in the adverse outcomes of assisted reproductive technology (ART), but little is known about the effects of ART on it. Primary ART techniques, including superovulation, in vitro fertilization (IVF), and oocyte in vitro maturation (IVM), were analyzed in this study of the effects of ART on embryo quality and Grb10. METHODS: Embryo development rates were determined. Blastocyst cell number and global methylation were analyzed at the single-embryo level, together with Grb10 methylation and mRNA expression of the imprinted genes. RESULTS: Lower blastocyst cell number, higher genome and Grb10 CGI1 methylation, and variable mRNA expression were observed in the ART groups compared with the control group. Whether fertilization was in vivo or in vitro, the changes in the genome and Grb10 CGI1 methylation level and Grb10 and H19 expression were similar in the groups with superovulation and more significant than the IVM group. CONCLUSIONS: These results suggest that superovulation had a greater impact than IVF or IVM on the genome and Grb10 DNA methylation level, and Grb10 and H19 expression.


Subject(s)
Blastocyst/metabolism , Fertilization in Vitro/methods , GRB10 Adaptor Protein/metabolism , Oocytes/metabolism , Superovulation/physiology , Animals , Female , Fertilization in Vitro/adverse effects , In Vitro Oocyte Maturation Techniques/methods , Mice
15.
Reprod Fertil Dev ; 30(11): 1443-1453, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29769162

ABSTRACT

Serial cloning by somatic cell nuclear transfer (SCNT) is a critical tool for the expansion of precious transgenic lines or resetting the lifespan of primary transgenic cells for multiple genetic modifications. We successfully produced second-generation cloned goats using donor neonatal fibroblasts from first-generation clones. However, our attempts to produce any third-generation clones failed. SCNT efficiency decreased progressively with the clonal generations. The rate of pregnancy loss was significantly greater in recloning groups (P<0.05). While no pregnancy loss was observed during the first round of SCNT, 14 out of 21 pregnancies aborted in the second round of SCNT and all pregnancies aborted in the third round of SCNT. In this retrospective study, we also investigated the expression of 21 developmentally important genes in muscle tissue of cloned (G1) and recloned (G2) offspring. The expression of most of these genes in live clones was found to be largely comparable to naturally reproduced control goats, but fibroblast growth factor 10 (FGF10), methyl CpG binding protein 2 (MECP2) and growth factor receptor bound protein 10 (GRB10) were differentially expressed (P<0.05) in G2 goats compared with G1 and controls. To study the effects of serial cloning on DNA methylation, the methylation pattern of differentially methylated regions in imprinted genes H19 and insulin like growth factor 2 receptor (IGF2R) were also analysed. Aberrant H19 DNA methylation patterns were detected in G1 and G2 clones.


Subject(s)
Abortion, Veterinary , Cloning, Organism/veterinary , DNA Methylation , Nuclear Transfer Techniques/veterinary , Animals , Animals, Genetically Modified , Female , Fibroblast Growth Factor 10/genetics , Fibroblast Growth Factor 10/metabolism , GRB10 Adaptor Protein/genetics , GRB10 Adaptor Protein/metabolism , Genomic Imprinting , Goats , Methyl-CpG-Binding Protein 2/genetics , Methyl-CpG-Binding Protein 2/metabolism , Muscle, Skeletal/metabolism , Pregnancy , Retrospective Studies
16.
Genetics ; 209(1): 233-239, 2018 05.
Article in English | MEDLINE | ID: mdl-29563147

ABSTRACT

Imprinted genes are expressed from one parental allele only as a consequence of epigenetic events that take place in the mammalian germ line and are thought to have evolved through intragenomic conflict between parental alleles. We demonstrate, for the first time, oppositional effects of imprinted genes on brain and behavior. Specifically, we show that mice lacking paternal Grb10 make fewer impulsive choices, with no dissociable effects on a separate measure of impulsive action. Taken together with previous work showing that mice lacking maternal Nesp55 make more impulsive choices, this suggests that impulsive choice behavior is a substrate for the action of genomic imprinting. Moreover, the contrasting effect of these two genes suggests that impulsive choices are subject to intragenomic conflict and that maternal and paternal interests pull this behavior in opposite directions. Finally, these data may also indicate that an imbalance in expression of imprinted genes contributes to pathological conditions such as gambling and drug addiction, where impulsive behavior becomes maladaptive.


Subject(s)
Behavior, Animal , GRB10 Adaptor Protein/genetics , Gene Expression , Genomic Imprinting , Analysis of Variance , Animals , Fluorescent Antibody Technique , GRB10 Adaptor Protein/metabolism , Immunohistochemistry , Impulsive Behavior , Male , Mice
17.
Eur Urol ; 73(6): 949-960, 2018 06.
Article in English | MEDLINE | ID: mdl-29544736

ABSTRACT

BACKGROUND: Although androgen deprivation therapy is initially effective in controlling growth of hormone-naive prostate cancers (HNPCs) in patients, currently incurable castration-resistant prostate cancer (CRPC) inevitably develops. OBJECTIVE: To identify CRPC driver genes that may provide new targets to enhance CRPC therapy. DESIGN, SETTING, AND PARTICIPANTS: Patient-derived xenografts (PDXs) of HNPCs that develop CRPC following host castration were examined for changes in expression of genes at various time points after castration using transcriptome profiling analysis; particular attention was given to pre-CRPC changes in expression indicative of genes acting as potential CRPC drivers. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS: The functionality of a potential CRPC driver was validated via its knockdown in cultured prostate cancer cells; its clinical relevance was established using data from prostate cancer patient databases. RESULTS AND LIMITATIONS: Eighty genes were found to be significantly upregulated at the CRPC stage, while seven of them also showed elevated expression prior to CRPC development. Among the latter, growth factor receptor bound protein 10 (GRB10) was the most significantly and consistently upregulated gene. Moreover, elevated GRB10 expression in clinical prostate cancer samples correlated with more aggressive tumor types and poorer patient treatment outcome. GRB10 knockdown markedly reduced prostate cancer cell proliferation and activity of AKT, a well-established CRPC mediator. A positive correlation between AKT activity and GRB10 expression was also found in clinical cohorts. CONCLUSIONS: GRB10 acts as a driver of CRPC and sensitizes androgen receptor pathway inhibitors, and hence GRB10 targeting provides a novel therapeutic strategy for the disease. PATIENT SUMMARY: Development of castration-resistant prostate cancer (CRPC) is a major problem in the management of the disease. Using state-of-the-art patient-derived hormone-naive prostate cancer xenograft models, we found and validated the growth factor receptor bound protein 10 gene as a driver of CRPC, indicating that it may be used as a new molecular target to enhance current CRPC therapy.


Subject(s)
GRB10 Adaptor Protein/genetics , Gene Expression , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , RNA, Messenger/metabolism , Receptors, Androgen/genetics , Animals , Castration , Cell Line, Tumor , Cell Proliferation/genetics , Disease Models, Animal , GRB10 Adaptor Protein/metabolism , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Heterografts , Humans , Male , Mice , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms, Castration-Resistant/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , Transcriptome , Up-Regulation
18.
J Mol Endocrinol ; 60(4): 285-297, 2018 05.
Article in English | MEDLINE | ID: mdl-29555819

ABSTRACT

The growth factor receptor bound protein GRB10 is an imprinted gene product and a key negative regulator of the insulin, IGF1 and mTORC1 signaling pathways. GRB10 is highly expressed in mouse fetal liver but almost completely silenced in adult mice, suggesting a potential detrimental role of this protein in adult liver function. Here we show that the Grb10 gene could be reactivated in adult mouse liver by acute endoplasmic reticulum stress (ER stress) such as tunicamycin or a short-term high-fat diet (HFD) challenge, concurrently with increased unfolded protein response (UPR) and hepatosteatosis. Lipogenic gene expression and acute ER stress-induced hepatosteatosis were significantly suppressed in the liver of the liver-specific GRB10 knockout mice, uncovering a key role of Grb10 reactivation in acute ER stress-induced hepatic lipid dysregulation. Mechanically, acute ER stress induces Grb10 reactivation via an ATF4-mediated increase in Grb10 gene transcription. Our study demonstrates for the first time that the silenced Grb10 gene can be reactivated by acute ER stress and its reactivation plays an important role in the early development of hepatic steatosis.


Subject(s)
Endoplasmic Reticulum Stress , Fatty Liver/metabolism , Fatty Liver/pathology , GRB10 Adaptor Protein/metabolism , Gene Silencing , Liver/metabolism , Liver/pathology , Activating Transcription Factor 4/metabolism , Aging/metabolism , Animals , Diet, High-Fat , Endoplasmic Reticulum Stress/drug effects , Fatty Acids/biosynthesis , Fatty Liver/genetics , Feeding Behavior , GRB10 Adaptor Protein/genetics , Gene Deletion , Gene Expression Regulation/drug effects , Gene Silencing/drug effects , Lipid Metabolism/drug effects , Liver/drug effects , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity/drug effects , Tunicamycin/pharmacology
19.
Cryobiology ; 80: 89-95, 2018 02.
Article in English | MEDLINE | ID: mdl-29180273

ABSTRACT

Transplantation of cryopreserved ovarian tissue has been considered as a promising way of fertility preservation for women. however, this cryopreservation method is prone to post-resuscitation follicle proliferation and oocyte development stagnation, affecting late transplant survival. To evaluate current vitrification works, we investigated the critical pathway alternations in vitrified-warmed juvenile 10-day-old mouse ovary. We showed a significant decrease of protein kinase B (Akt) and Mitogen-activated protein kinase (Mapk) phosphorylation, during which serine/threonine kinases play central roles in coordinating follicle and oocyte development and stress response. Inhibition of Akt and Mapk activity were associated with one of the imprinted insulin pathway negative regulatory genes, Growth factor receptor-binding protein 10 (Grb10) which remarkably increased in vitrified-warmed juvenile mouse ovary than that of fresh group (p < 0.05). RNAi-induced Grb10 down-regulation reversed the decrease in Akt and Mapk phosphorylation. The increase of Grb10 expression was partially caused by the hyper-methylation of the promoter region, associated with the decrease of follicular DNA methyltransferase (Dnmt) 1 protein in different stages of vitrified-warmed group, compared to fresh group (p < 0.05). The mRNA and protein expression of Dnmt1 in ovary of vitrified-warmed juvenile mouse were remarkably lower than those in fresh group (p < 0.05). Dnmt1 overexpression dramatically reversed Grb10 up-regulation and Akt and Mapk phosphorylation reduction. Taken together, our findings suggest that Grb10 expression might be helpful in evaluation of effectiveness of vitrification, and considered as a potential target for further vitrification protocols improvement in the future.


Subject(s)
Cryopreservation/methods , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , GRB10 Adaptor Protein/metabolism , Ovarian Follicle/metabolism , Vitrification , Animals , DNA/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA Methylation , Female , Fertility Preservation/methods , GRB10 Adaptor Protein/genetics , Humans , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Oocytes/cytology , Ovarian Follicle/cytology , Ovarian Follicle/transplantation , Phosphorylation , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering/genetics
20.
Nutrients ; 9(11)2017 Oct 27.
Article in English | MEDLINE | ID: mdl-29077002

ABSTRACT

The mammalian target of rapamycin (mTOR) is a serine/threonine kinase that controls a wide spectrum of cellular processes, including cell growth, differentiation, and metabolism. mTOR forms two distinct multiprotein complexes known as mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2), which are characterized by the presence of raptor and rictor, respectively. mTOR controls insulin signaling by regulating several downstream components such as growth factor receptor-bound protein 10 (Grb10), insulin receptor substrate (IRS-1), F-box/WD repeat-containing protein 8 (Fbw8), and insulin like growth factor 1 receptor/insulin receptor (IGF-IR/IR). In addition, mTORC1 and mTORC2 regulate each other through a feedback loop to control cell growth. This review outlines the current understanding of mTOR regulation in insulin signaling in the context of whole body metabolism.


Subject(s)
Insulin/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Proliferation , F-Box Proteins/metabolism , GRB10 Adaptor Protein/metabolism , Humans , Insulin Receptor Substrate Proteins/metabolism , Rapamycin-Insensitive Companion of mTOR Protein/metabolism , Receptor, IGF Type 1/metabolism , Receptor, Insulin/metabolism , Regulatory-Associated Protein of mTOR/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...