Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 361
Filter
1.
Nat Commun ; 15(1): 1007, 2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38307855

ABSTRACT

Proper cellular proteostasis, essential for viability, requires a network of chaperones and cochaperones. ATP-dependent chaperonin TRiC/CCT partners with cochaperones prefoldin (PFD) and phosducin-like proteins (PhLPs) to facilitate folding of essential eukaryotic proteins. Using cryoEM and biochemical analyses, we determine the ATP-driven cycle of TRiC-PFD-PhLP2A interaction. PhLP2A binds to open apo-TRiC through polyvalent domain-specific contacts with its chamber's equatorial and apical regions. PhLP2A N-terminal H3-domain binding to subunits CCT3/4 apical domains displace PFD from TRiC. ATP-induced TRiC closure rearranges the contacts of PhLP2A domains within the closed chamber. In the presence of substrate, actin and PhLP2A segregate into opposing chambers, each binding to positively charged inner surface residues from CCT1/3/6/8. Notably, actin induces a conformational change in PhLP2A, causing its N-terminal helices to extend across the inter-ring interface to directly contact a hydrophobic groove in actin. Our findings reveal an ATP-driven PhLP2A structural rearrangement cycle within the TRiC chamber to facilitate folding.


Subject(s)
Actins , Eye Proteins , GTP-Binding Protein Regulators , Phosphoproteins , Protein Folding , Actins/metabolism , Carrier Proteins/metabolism , Chaperonins/metabolism , Adenosine Triphosphate/metabolism , Chaperonin Containing TCP-1/metabolism
2.
Curr Cancer Drug Targets ; 23(1): 71-86, 2022.
Article in English | MEDLINE | ID: mdl-35708078

ABSTRACT

OBJECTIVE: Identification and validation of genes that functionally account for the growth and metastasis of prostate cancer. METHODS: DU145-KO cell line was constructed by transfecting DU145 cells with lentivirus packaged with the genome-wide knock-out library. The DU145-KO cells were transplanted into the armpits of immunocompromised Nu/Nu mice, followed by the tissue collection from the lung at week 3 (early lung tissue) or week 7 (late lung tissue with micro-metastasis), as well as from primary tumor site at week 7 (late primary tumor) after inoculation. Lung metastasis was retrieved at various time points for DNA sequencing analysis to identify enriched sgRNAs, thus candidate genes/miRNAs. Further bioinformatics analysis and limited functional validation studies were carried out. RESULTS: DU145-KO cells promoted the formation of transplanted tumors in mice and promoted the growth and metastasis of primary tumors, compared to the controls (DU145-NC cells). The analysis of sequence data showed that the abundance of sgRNAs significantly changed in the primary tumor and micro-metastasis site. Fifteen target genes (C1QTNF9B, FAM229A, hsa-mir-3929, KRT23, TARS2, CRADD, GRIK4, PLA2G15, LOXL1, SLITRK6, CDC42EP5, SLC2A4, PTGDS, MYL9 and ACOX2 for the enriched sgRNAs) have been selected for experimental validation, which showed that knock-out of any of these genes led to the enhanced potential of invasion and metastasis of DU145 cells. CONCLUSION: Genome-wide CRISPR-Cas9 knock-out screening technology combined with highthroughput sequencing analysis identified genes that potentially relate to prostate tumor invasion and metastasis. Analysis of these genes provided insights into biological pathways relevant to the disease and disclosed innovative markers for diagnosis or prognosis as well as potential targets for therapy.


Subject(s)
MicroRNAs , Prostatic Neoplasms , Humans , Male , Mice , Animals , CRISPR-Cas Systems , Cell Line, Tumor , Early Detection of Cancer , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , MicroRNAs/genetics , GTP-Binding Protein Regulators/genetics
3.
Cell Death Dis ; 13(4): 290, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35365622

ABSTRACT

Gliomas are the most common brain malignancies characterized by high degree of aggressiveness and high mortality. However, the underlying mechanism of glioma progression remains unclear. Here, we probed the role of CDC42EP3 (CDC42 effector protein 3) played in glioma development and its potential downstream mechanism. The expression of CDC42EP3 in tumor and normal brain tissues were examined through immunohistochemistry and we found the likelihood of CDC42EP3 overexpression was positively correlated with pathological grading. Patients with higher expression of CDC42EP3 were more likely to suffer from recurrence as well. Through constructing CDC42EP3-knockdown cell models, we discovered that silencing CDC42EP3 significantly restricted cell proliferation and migration but facilitated cell apoptosis in vitro. Inhibition on tumor growth mediated by CDC42EP3 depletion was further verified in vivo. Regarding downstream target of CDC42EP3, we found that it may positively regulate the expression of CCND1 through c-Myc-mediated transcription. Furthermore, our findings affirmed that effects of CDC42EP3 overexpression on cell proliferation, migration and apoptosis could be confined by depleting CCND1. In a word, this study reported the tumor-promoting role of CDC42EP3 in glioma progression which probably functioned through targeting CCND1.


Subject(s)
Brain Neoplasms , Glioma , Apoptosis/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin D1/genetics , Cyclin D1/metabolism , GTP-Binding Protein Regulators/genetics , GTP-Binding Protein Regulators/metabolism , Gene Expression Regulation, Neoplastic , Glioma/pathology , Humans
4.
Exp Cell Res ; 412(1): 113018, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34998812

ABSTRACT

BACKGROUND: Osteosarcoma is a disease with high mortality of malignant tumors in children and adolescents. CDC42 effector protein 3 (CDC42EP3) has been reported to be associated with human cancer cell progression. This study aimed to investigate the biological function and preliminary molecular mechanism of CDC42EP3 in osteosarcoma. METHODS: CDC42EP3 expression in osteosarcoma was analyzed by immunohistochemical (IHC) staining. Secondly, the biological effects of CDC42EP3 in osteosarcoma cells was determined by loss/gain-of-function assays in vitro and in vivo. RESULTS: CDC42EP3 expression was higher in osteosarcoma tissue than in noncancerous tissue. The expression of CDC42EP3 was positively correlated with age, pathological stage and grade of patients with osteosarcoma. Furthermore, downregulation of CDC42EP3 suppressed tumor progression by inhibiting proliferation, migration and inducing apoptosis in vivo. Importantly, knockdown of CDC42EP3 reduced the expression of interstitial markers (N-cadherin, Vimentin and Snail) and increased the expression of epithelial markers (E-cadherin). In addition, CDC42EP3 knockdown downregulated PI3K and reduced the phosphorylation levels of AKT and mTOR. The mice xenograft model further confirmed that CDC42EP3 knockdown inhibited osteosarcoma growth in vitro. CONCLUSIONS: In summary, these findings highlighted the significance of CDC42EP3 in tumor progression, which implicated CDC42EP3 as a promising candidate molecular target for osteosarcoma therapy.


Subject(s)
Bone Neoplasms/metabolism , GTP-Binding Protein Regulators/metabolism , Osteosarcoma/metabolism , Adult , Animals , Apoptosis , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cell Survival , Disease Progression , Female , GTP-Binding Protein Regulators/antagonists & inhibitors , GTP-Binding Protein Regulators/genetics , Gene Knockdown Techniques , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Osteosarcoma/genetics , Osteosarcoma/pathology
5.
Mol Med ; 28(1): 3, 2022 01 14.
Article in English | MEDLINE | ID: mdl-35030992

ABSTRACT

The mechanisms of chronic intermittent hypoxia (CIH)-induced cognitive deficits remain unclear. Here, our study found that about 3 months CIH treatment induced lipid droplets (LDs) accumulation in hippocampal nerve and glia cells of C57BL/6 mice, and caused severe neuro damage including neuron lesions, neuroblast (NB) apoptosis and abnormal glial activation. Studies have shown that the neuronal metabolism disorders might contribute to the CIH induced-hippocampal impairment. Mechanistically, the results showed that pyruvate dehydrogenase complex E1ɑ subunit (PDHA1) and the pyruvate dehydrogenase complex (PDC) activator pyruvate dehydrogenase phosphatase 1 (PDP1) did not noticeable change after intermittent hypoxia. Consistent with those results, the level of Acetyl-CoA in hippocampus did not significantly change after CIH exposure. Interestingly, we found that CIH produced large quantities of ROS, which activated the JNK/SREBP/ACC pathway in nerve and glia cells. ACC catalyzed the carboxylation of Acetyl-CoA to malonyl-CoA and then more lipid acids were synthesized, which finally caused aberrant LDs accumulation. Therefore, the JNK/SREBP/ACC pathway played a crucial role in the cognitive deficits caused by LDs accumulation after CIH exposure. Additionally, LDs were peroxidized by the high level of ROS under CIH conditions. Together, lipid metabolic disorders contributed to nerve and glia cells damage, which ultimately caused behavioral dysfunction. An active component of Salvia miltiorrhiza, SMND-309, dramatically alleviated these injuries and improved cognitive deficits of CIH mice.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Cognition , Eye Proteins/metabolism , GTP-Binding Protein Regulators/metabolism , Lipid Droplets/metabolism , Phosphoproteins/metabolism , Protein Phosphatase 2C/metabolism , Sleep Apnea, Obstructive/etiology , Sleep Apnea, Obstructive/metabolism , Sterol Regulatory Element Binding Proteins/metabolism , Animals , Caffeic Acids/pharmacology , Cognitive Dysfunction , Disease Models, Animal , Hippocampus/metabolism , Hypoxia/metabolism , Learning , Lipid Peroxidation , MAP Kinase Signaling System , Memory , Mice , Neurons , Reactive Oxygen Species/metabolism , Signal Transduction , Sleep Apnea, Obstructive/diagnosis , Sleep Apnea, Obstructive/drug therapy
6.
Blood ; 138(17): 1628-1636, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34269803

ABSTRACT

Human cytomegalovirus (CMV) reactivation is a frequent complication of allogeneic hematopoietic cell transplantation (HCT). Despite routine screening for CMV reactivation and early antiviral treatment, the rates of CMV-related complications after HCT remain high. Genetic variants in both the donor and recipient have been associated with the risk of CMV reactivation and disease after HCT, but these associations have not been validated, and their clinical importance remains unclear. In this study, we assessed 117 candidate variants previously associated with CMV-related phenotypes for association with CMV reactivation and disease in a cohort of 2169 CMV-seropositive HCT recipients. We also carried out a genome-wide association study (GWAS) for CMV reactivation and disease in the same cohort. Both analyses used a prespecified discovery and replication approach to control the risk of false-positive results. Among the 117 candidate variants, our analysis implicates only the donor ABCB1 rs1045642 genotype as a risk factor for CMV reactivation. This synonymous variant in P-glycoprotein may influence the risk of CMV reactivation by altering the efflux of cyclosporine and tacrolimus from donor lymphocytes. In the GWAS analysis, the donor CDC42EP3 rs11686168 genotype approached the significance threshold for association with CMV reactivation, although we could not identify a mechanism to explain this association. The results of this study suggest that most genomic variants previously associated with CMV phenotypes do not significantly alter the risk for CMV reactivation or disease after HCT.


Subject(s)
Cytomegalovirus Infections/genetics , Hematopoietic Stem Cell Transplantation/adverse effects , ATP Binding Cassette Transporter, Subfamily B/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Cytomegalovirus/isolation & purification , Cytomegalovirus/physiology , Cytomegalovirus Infections/etiology , Female , GTP-Binding Protein Regulators/genetics , Genome-Wide Association Study , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Transplantation, Homologous/adverse effects , Virus Activation , Young Adult
7.
Curr Biol ; 31(18): 4088-4103.e5, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34329591

ABSTRACT

Cell resistance to taxanes involves several complementary mechanisms, among which septin relocalization from actin stress fibers to microtubules plays an early role. By investigating the molecular mechanism underlying this relocalization, we found that acute paclitaxel treatment triggers the release from stress fibers and subsequent proteasome-mediated degradation of binder of Rho GTPases 2 (BORG2)/Cdc42 effector protein 3 (Cdc42EP3) and to a lesser extent of BORG3/Cdc42EP5, two Cdc42 effectors that link septins to actin in interphase cells. BORG2 or BORG3 silencing not only caused septin detachment from stress fibers but also mimicked the effects of paclitaxel by triggering both septin relocalization to microtubules and significant drug resistance. Conversely, BORG2 or BORG3 overexpression retained septins on actin fibers even after paclitaxel treatment, without affecting paclitaxel sensitivity. We found that drug-induced inhibition of Cdc42 resulted in a drop in BORG2 level and in the relocalization of septins to microtubules. Accordingly, although septins relocalized when overexpressing an inactive mutant of Cdc42, the expression of a constitutively active mutant acted locally at actin stress fibers to prevent septin release, even after paclitaxel treatment. These findings reveal the role of Cdc42 upstream of BORG2 and BORG3 in controlling the interplay between septins, actin fibers, and microtubules in basal condition and in response to taxanes.


Subject(s)
Actins , Septins , Actins/metabolism , GTP-Binding Protein Regulators/metabolism , Microtubules/metabolism , Septins/genetics , Septins/metabolism , Stress Fibers/metabolism
8.
Carcinogenesis ; 42(9): 1179-1188, 2021 10 05.
Article in English | MEDLINE | ID: mdl-34111280

ABSTRACT

Gastric cancer (GC) is one of the most prevalent cancers and severely endangers human health. Due to the low rate of diagnosis, most patients with GC are diagnosed as advanced. CDC42 effector protein 3 (CDC42EP3) has been revealed to be involved in several types of human cancers' development and progression. However, the function of CDC42EP3 in GC is not yet clear. CDC42EP3 expression was detected by immunohistochemistry, quantitative real-time PCR and Western blot assay in tumor tissues and cell lines of GC. CDC42EP3 knockdown cell models were constructed by lentivirus transfection. Cell proliferation was evaluated by the 3-(4,5-dimethylthiazole-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The wound-healing assay and the transwell assay were utilized to assess the cell migration. Also, the cell apoptosis and the cell cycle were evaluated by flow cytometry. Moreover, the mechanism was investigated by Human Apoptosis Antibody Array. The in vivo experiments were conducted to verify the effects of CDC42EP3 knockdown on the tumor growth of GC. The expression level of CDC42EP3 was up-regulated in tumor tissues. High CDC42EP3 expression was positively related to more advanced tumor grade. CDC42EP3 knockdown inhibited cell proliferation and migration, promoted cell apoptosis and suppressed the tumor growth. On the other hand, it was also found that the silencing of CDC42EP3 inhibited HSP27 and IGF-1sR expression as well as promoted Caspase3, p53, TNF-α, TNF-ß, TRAILR-1 and TRAILR-2 expression. CDC42EP3 was revealed to work as a tumor promoter in the development and progression of GC, which could be a promising therapeutic target for the therapy of GC.


Subject(s)
GTP-Binding Protein Regulators/metabolism , Stomach Neoplasms , Aged , Animals , Apoptosis , Cell Movement , Cell Proliferation , Disease Progression , Female , GTP-Binding Protein Regulators/genetics , Gene Knockdown Techniques , Gene Silencing , Heterografts , Humans , Male , Mice , Middle Aged , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Survival Rate , Up-Regulation
9.
IUBMB Life ; 73(9): 1103-1114, 2021 09.
Article in English | MEDLINE | ID: mdl-34060227

ABSTRACT

This study was aimed at determining the roles and functions of lncRNA XIST/miR-545-3p/G3BP2 axis during hypoxia/reoxygenation (H/R)-induced H9C2 cell apoptosis. H9C2 cells were distributed into two groups, the H/R injury and control groups. High-throughput lncRNA sequencing was applied in the determination of differentially expressed lncRNAs between H/R-induced H9C2 cells and normal H9C2 cells. Real-time polymerase chain reactions (RT-PCR) were used to confirm the expression levels of lncRNA XIST in H/R-induced H9C2 cells. H9C2 cells were then transfected with lncRNA XIST recombinant plasmid (lncRNA XIST), sh-LINC XIST, agomiR-545-3p, antagomiR-545-3p, pcDNA-G3BP2, sh-G3BP2, and a corresponding negative control (NC). Bioinformatic analyses revealed that MiR-545-3p was a target for lncRNA XIST. This finding was confirmed by dual-luciferase reporter assay. The degree of cell apoptosis was evaluated by a flow cytometer. RT-PCR and western blot were performed to assess the apoptotic-related proteins in each group. A total of 859 differentially expressed lncRNAs (up-regulated = 502, down-regulated = 357) were identified. LncRNA XIST was found to be down-regulated in H/R-induced H9C2 cells while miR-545-3p was distinctly up-regulated. miR-545-3p was established to be a direct target for LncRNA XIST. LncRNA XIST significantly enhanced the apoptotic rate, while its inhibition suppressed the apoptotic rate. AgomiR-545-3p partially blocked the lncRNA XIST and enhanced the apoptosis of H/R-induced H9C2 cells. Moreover, miR-545-3p was shown to be a direct target for G3BP2. The overexpression of G3BP2 partially reversed the apoptotic effects of miR-545-3p on H/R-induced H9C2 cells. lncRNA XIST/miR-545-3p/GBP2 was found to be an apoptotic regulator in H/R-induced H9C2 cells.


Subject(s)
Apoptosis , Cell Hypoxia , GTP-Binding Protein Regulators , Myocytes, Cardiac , RNA, Long Noncoding , Animals , Male , Apoptosis/genetics , Cell Hypoxia/genetics , Gene Expression Regulation , Gene Knockdown Techniques , GTP-Binding Protein Regulators/genetics , GTP-Binding Protein Regulators/metabolism , MicroRNAs/genetics , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/pathology , Myocytes, Cardiac/physiology , Oxygen/metabolism , Rats, Sprague-Dawley , RNA, Long Noncoding/genetics
10.
Cell ; 183(2): 503-521.e19, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33007266

ABSTRACT

The control over the extent and timing of G protein signaling is provided by the regulator of G protein signaling (RGS) proteins that deactivate G protein α subunits (Gα). Mammalian genomes encode 20 canonical RGS and 16 Gα genes with key roles in physiology and disease. To understand the principles governing the selectivity of Gα regulation by RGS, we examine the catalytic activity of all canonical human RGS proteins and their selectivity for a complete set of Gα substrates using real-time kinetic measurements in living cells. The data reveal rules governing RGS-Gα recognition, the structural basis of its selectivity, and provide principles for engineering RGS proteins with defined selectivity. The study also explores the evolution of RGS-Gα selectivity through ancestral reconstruction and demonstrates how naturally occurring non-synonymous variants in RGS alter signaling. These results provide a blueprint for decoding signaling selectivity and advance our understanding of molecular recognition principles.


Subject(s)
GTP-Binding Protein alpha Subunits/metabolism , GTP-Binding Protein alpha Subunits/physiology , RGS Proteins/genetics , Animals , Female , GTP-Binding Protein Regulators/metabolism , GTP-Binding Protein alpha Subunits/genetics , HEK293 Cells , Humans , Kinetics , Male , Mice , Mice, Inbred C57BL , Neurons/metabolism , Primary Cell Culture , Protein Binding , RGS Proteins/metabolism , RGS Proteins/physiology , Signal Transduction/genetics
11.
J Cell Biol ; 219(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32798219

ABSTRACT

Fast amoeboid migration is critical for developmental processes and can be hijacked by cancer cells to enhance metastatic dissemination. This migratory behavior is tightly controlled by high levels of actomyosin contractility, but how it is coupled to other cytoskeletal components is poorly understood. Septins are increasingly recognized as novel cytoskeletal components, but details on their regulation and contribution to migration are lacking. Here, we show that the septin regulator Cdc42EP5 is consistently required for amoeboid melanoma cells to invade and migrate into collagen-rich matrices and locally invade and disseminate in vivo. Cdc42EP5 associates with actin structures, leading to increased actomyosin contractility and amoeboid migration. Cdc42EP5 affects these functions through SEPT9-dependent F-actin cross-linking, which enables the generation of F-actin bundles required for the sustained stabilization of highly contractile actomyosin structures. This study provides evidence that Cdc42EP5 is a regulator of cancer cell motility that coordinates actin and septin networks and describes a unique role for SEPT9 in melanoma invasion and metastasis.


Subject(s)
Actomyosin/metabolism , Cell Movement/physiology , GTP-Binding Protein Regulators/metabolism , Septins/metabolism , Actins/metabolism , Animals , Cell Line, Tumor , Cytoskeleton/metabolism , Focal Adhesions/metabolism , Humans , Melanoma/metabolism , Mice , Skin Neoplasms/metabolism , Melanoma, Cutaneous Malignant
12.
Exp Cell Res ; 395(2): 112206, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32739212

ABSTRACT

The small GTPase Cdc42, a member of the Rho family, regulates essential biological processes such as cytoskeleton remodeling, migration, vesicular trafficking and cell cycle. It was demonstrated that Cdc42 overactivation through different molecular strategies increases cell sensitivity to genotoxic stress and affects the phosphorylation status of DNA damage response proteins by unknown mechanisms. By using a combination of approaches including affinity purification/mass spectrometry (AP/MS) and colocalization microscopy analysis we were able to identify Cdc42EP3/Borg2 as a putative molecular effector of these molecular and cellular events that seem to be independent of cell line or DNA damage stimuli. We then investigated the influence of Cdc42EP3/Borg2 and other potential protein partners, such as the NCK and Septin2 proteins, which could mediate cellular responses to genotoxic stress under different backgrounds of Cdc42 activity. Clonogenic assays showed a reduced cell survival when ectopically expressing the Cdc42EP3/Borg2, NCK2 or Septin2 in an overactivated Cdc42-dependent background. Moreover, endogenous NCK appears to relocate into the nucleus upon Cdc42 overactivation, especially under genotoxic stress, and promotes the suppression of Chk1 phosphorylation. In sum, our findings reinforce Cdc42 as an important player involved in the DNA damage response acting through Cdc42EP3/Borg2 and NCK proteins following genomic instability conditions.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DNA/metabolism , GTP-Binding Protein Regulators/metabolism , Oncogene Proteins/metabolism , cdc42 GTP-Binding Protein/metabolism , Cell Movement/physiology , Cell Survival/physiology , Cytoskeleton/metabolism , Genomic Instability/genetics , Signal Transduction/physiology
13.
Proc Natl Acad Sci U S A ; 117(12): 6540-6549, 2020 03 24.
Article in English | MEDLINE | ID: mdl-32161136

ABSTRACT

The eukaryotic endomembrane system is controlled by small GTPases of the Rab family, which are activated at defined times and locations in a switch-like manner. While this switch is well understood for an individual protein, how regulatory networks produce intracellular activity patterns is currently not known. Here, we combine in vitro reconstitution experiments with computational modeling to study a minimal Rab5 activation network. We find that the molecular interactions in this system give rise to a positive feedback and bistable collective switching of Rab5. Furthermore, we find that switching near the critical point is intrinsically stochastic and provide evidence that controlling the inactive population of Rab5 on the membrane can shape the network response. Notably, we demonstrate that collective switching can spread on the membrane surface as a traveling wave of Rab5 activation. Together, our findings reveal how biochemical signaling networks control vesicle trafficking pathways and how their nonequilibrium properties define the spatiotemporal organization of the cell.


Subject(s)
Intracellular Membranes/metabolism , rab5 GTP-Binding Proteins/metabolism , Feedback, Physiological , GTP-Binding Protein Regulators/metabolism , Guanosine Diphosphate/metabolism , Intracellular Membranes/chemistry , Models, Biological , Protein Prenylation , Protein Transport , Signal Transduction , Stochastic Processes , Vesicular Transport Proteins/metabolism , rab5 GTP-Binding Proteins/chemistry
14.
J Appl Toxicol ; 40(7): 908-917, 2020 07.
Article in English | MEDLINE | ID: mdl-32077112

ABSTRACT

Pharmaceuticals are widely detected in aquatic environments, and their potential risks to aquatic species are of concern because they are designed to be biologically active. Here, we used an in vitro assay, called the transforming growth factor α shedding assay, to measure the biological activities of G protein-coupled receptor (GPCR)-acting pharmaceuticals present in river water and effluents from municipal wastewater treatment plants (WWTPs) in Japan from 2014 to 2016. Antagonistic activities against angiotensin (AT1), dopamine (D2), adrenergic (ß1), acetylcholine (M1) and histamine (H1) receptors were detected in river water, and were stronger downstream than upstream owing to effluent from WWTPs along the river. Ozonation at one WWTP reduced these activities. Concentrations of sulpiride (D2 antagonist) could explain 73% of antagonistic activities against the D2 receptor; those of metoprolol, atenolol and propranolol (ß1 antagonists) could explain 16% of activities against the ß1 receptor; and those of pirenzepine (M1 antagonist) could explain 15% of activities against the M1 receptor. Therefore, other receptor antagonists also occur. GPCR-acting pharmaceuticals should be given more attention in environmental monitoring and toxicity testing.


Subject(s)
GTP-Binding Protein Regulators/toxicity , Pharmaceutical Preparations/analysis , Pharmaceutical Preparations/chemistry , Wastewater/chemistry , Wastewater/toxicity , Water Pollutants, Chemical/analysis , Water Pollutants, Chemical/toxicity , Drug-Related Side Effects and Adverse Reactions , Environmental Monitoring , Fresh Water/chemistry , GTP-Binding Protein Regulators/analysis , Japan , Rivers/chemistry
15.
Nat Commun ; 10(1): 3779, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31439829

ABSTRACT

Evolution is proposed to result, in part, from acquisition of new developmental programs. One such example is the appearance of the micromeres in a sea urchin that form by an asymmetric cell division at the 4th embryonic cleavage and function as a major signaling center in the embryo. Micromeres are not present in other echinoderms and thus are  considered as a derived feature, yet its acquisition mechanism is unknown. Here, we report that the polarity factor AGS and its associated proteins are responsible for micromere formation. Evolutionary modifications of AGS protein seem to have provided the cortical recruitment and binding of AGS to the vegetal cortex, contributing to formation of micromeres in the sea urchins. Indeed, introduction of sea urchin AGS into the sea star embryo induces asymmetric cell divisions, suggesting that the molecular evolution of AGS protein is key in the transition of echinoderms to micromere formation and the current developmental style of sea urchins not seen in other echinoderms.


Subject(s)
Blastomeres/physiology , Cell Division/physiology , Embryo, Nonmammalian/embryology , GTP-Binding Protein Regulators/metabolism , Sea Urchins/embryology , Animals , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Sea Urchins/metabolism , Signal Transduction/genetics
16.
Fungal Biol ; 122(6): 479-486, 2018 06.
Article in English | MEDLINE | ID: mdl-29801792

ABSTRACT

Trichoderma reesei represents one of the most prolific producers of homologous and heterologous proteins. Discovery of the photoreceptor ENV1 as a regulator of cellulase gene expression initiated analysis of light response pathways and their physiological relevance for T. reesei. The function of ENV1 in regulation of plant cell wall degrading enzymes is conserved in Neurospora crassa. ENV1 emerged as a central checkpoint for integration of nutrient sensing, light response and development. This photoreceptor exerts its function by influencing transcript abundance and feedback cycles of the alpha subunits of the heterotrimeric G-protein pathway and impacts regulation of the beta and gamma subunits via mutual regulation with the phosducin PhLP1. The output of regulation by ENV1 is in part mediated by the cAMP pathway and likely aimed at cellulose recognition. Lack of ENV1 causes deregulation of the pheromone system and female sterility in light. A regulatory interconnection with VEL1 and influence on other regulators of secondary metabolism like YPR2 as well as polyketide synthase encoding genes indicates a function in secondary metabolism. The function of ENV1 in integrating light response with signaling of osmotic and oxidative stress is evolutionary conserved in Hypocreales and distinct from other sordariomycetes including N. crassa.


Subject(s)
Fungal Proteins/metabolism , Photoreceptors, Microbial/metabolism , Trichoderma/physiology , Eye Proteins/metabolism , Fungal Proteins/genetics , GTP-Binding Protein Regulators/metabolism , Gene Expression Regulation, Fungal , Heterotrimeric GTP-Binding Proteins/genetics , Pheromones/metabolism , Phosphoproteins/metabolism , Photoreceptors, Microbial/genetics , Secondary Metabolism , Signal Transduction , Trichoderma/genetics , Trichoderma/radiation effects
17.
Med Sci Monit ; 24: 2210-2217, 2018 Apr 13.
Article in English | MEDLINE | ID: mdl-29652036

ABSTRACT

BACKGROUND The cardiac autonomic nervous system plays a crucial role in genesis and development of atrial fibrillation (AF) through the G protein signal transduction pathway. Therefore, intervening in the G protein signal transduction pathway may be a new "selective drug" method to regulate autonomic nerve activity to prevent vagally-mediated AF. MATERIAL AND METHODS Seventeen adult beagles were randomized into 3 groups: shame-operation control group (group A, n=5), empty vector gene control group (group B, n=6), and Gαi2ctp gene experimental group (group C, n=6). Group A was injected with normal saline into the anterior atrial wall, and group B and group C animals were injected with recombinant adenovirus with empty vector or Gαi2ctp vector in the same region. AF was induced by the method of rapid atrial pacing in groups B and C. To determine the clinical effect of vagal modulation, the effective refractory periods (ERP) and field action potential duration (FAPD) were evaluated by electrophysiological study. The expression levels of tyrosine hydroxylase (TH) and choline acetyl transferase (CHAT) in different parts were determined with immunohistochemistry. RESULTS After successful Gai2ctp gene transfer, in group B, the ERP and FAPD significantly decreased (P<0.05), and TH and CHAT expression observably increased (P<0.05), while those differences were absent between groups A and C (P>0.05). CONCLUSIONS Recombinant adenovirus-mediated overexpression of Gαi2ctp in canine myocardial cells can interfere with the activity of the vagus nerve, reverse the development and progression of electrical remodeling, and reduce the incidence of AF.


Subject(s)
Atrial Fibrillation/physiopathology , GTP-Binding Protein Regulators/metabolism , Vagus Nerve/drug effects , Action Potentials , Animals , Atrial Fibrillation/metabolism , Atrial Remodeling , Autonomic Nervous System/physiopathology , Cardiac Pacing, Artificial/methods , Choline O-Acetyltransferase/analysis , Dogs , Heart Atria/physiopathology , Signal Transduction , Sympathetic Nervous System/physiopathology , Tyrosine 3-Monooxygenase/analysis , Vagus Nerve/physiology
18.
Int J Mol Sci ; 19(4)2018 Apr 11.
Article in English | MEDLINE | ID: mdl-29641463

ABSTRACT

Rhegmatogenous retinal detachment (RRD) is a potentially blinding condition characterized by a physical separation between neurosensory retina and retinal pigment epithelium. Quantitative proteomics can help to understand the changes that occur at the cellular level during RRD, providing additional information about the molecular mechanisms underlying its pathogenesis. In the present study, iTRAQ labeling was combined with two-dimensional LC-ESI-MS/MS to find expression changes in the proteome of vitreous from patients with RRD when compared to control samples. A total of 150 proteins were found differentially expressed in the vitreous of patients with RRD, including 96 overexpressed and 54 underexpressed. Several overexpressed proteins, several such as glycolytic enzymes (fructose-bisphosphate aldolase A, gamma-enolase, and phosphoglycerate kinase 1), glucose transporters (GLUT-1), growth factors (metalloproteinase inhibitor 1), and serine protease inhibitors (plasminogen activator inhibitor 1) are regulated by HIF-1, which suggests that HIF-1 signaling pathway can be triggered in response to RRD. Also, the accumulation of photoreceptor proteins, including phosducin, rhodopsin, and s-arrestin, and vimentin in vitreous may indicate that photoreceptor degeneration occurs in RRD. Also, the accumulation of photoreceptor proteins, including phosducin, rhodopsin, and s-arrestin, and vimentin in vitreous may indicate that photoreceptor degeneration occurs in RRD. Nevertheless, the differentially expressed proteins found in this study suggest that different mechanisms are activated after RRD to promote the survival of retinal cells through complex cellular responses.


Subject(s)
Proteome/genetics , Retinal Detachment/metabolism , Aged , Arrestin/genetics , Arrestin/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Female , GTP-Binding Protein Regulators/genetics , GTP-Binding Protein Regulators/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glycolysis , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Male , Middle Aged , Phosphoproteins/genetics , Phosphoproteins/metabolism , Proteome/metabolism , Retina/metabolism , Retinal Detachment/genetics , Rhodopsin/genetics , Rhodopsin/metabolism
19.
Acta Pharmacol Sin ; 39(2): 184-194, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28816235

ABSTRACT

The RasGAP SH3 domain-binding proteins (G3BPs) are a family of RNA-binding proteins that can co-ordinate signal transduction and post-transcriptional gene regulation. G3BPs have been shown to be involved in mediating a great diversity of cellular processes such as cell survival, growth, proliferation and apoptosis. But the potential roles of G3BPs in the pathogenesis and progression of cardiovascular diseases remain to be clarified. In the present study, we provide the first evidence that suggests the participation of G3BP2 in cardiac hypertrophy. In cultured neonatal rat cardiomyocytes (NRCMs), treatment with isoproterenol (ISO, 0.1-100 µmol/L) significantly elevated the mRNA and protein levels of G3BP2. Similar results were observed in the hearts of rats subjected to 7D-injection of ISO, accompanied by obvious heart hypertrophy and elevated the expression of hypertrophy marker genes ANF, BNP and ß-MHC in heart tissues. Overexpression of G3BP2 in NRCMs led to hypertrophic responses evidenced by increased cellular surface area and the expression of hypertrophy marker genes, whereas knockdown of G3BP2 significantly attenuated ISO-induced hypertrophy of NRCMs. We further showed that G3BP2 directly interacted with IκBα and promoted the aggregation of the NF-κB subunit p65 in the nucleus and increased NF-κB-dependent transcriptional activity. NF-κB inhibition with PDTC (50 µmol/L) or p65 knockdown significantly decreased the hypertrophic responses in NRCMs induced by ISO or G3BP2 overexpression. These results give new insight into the functions of G3BP2 and may help further elucidate the molecular mechanisms underlying cardiac hypertrophy.


Subject(s)
Cardiomegaly/metabolism , GTP-Binding Protein Regulators/metabolism , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , RNA-Binding Proteins/metabolism , Animals , Cardiomegaly/chemically induced , Cardiomegaly/pathology , Cell Nucleus/metabolism , Disease Models, Animal , GTP-Binding Protein Regulators/genetics , Gene Knockdown Techniques , Isoproterenol , Male , Myocytes, Cardiac/pathology , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/antagonists & inhibitors , Pyrrolidines/pharmacology , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Rats, Sprague-Dawley , Signal Transduction/drug effects , Thiocarbamates/pharmacology , Transcription Factor RelA/metabolism
20.
Biotechnol Lett ; 40(1): 57-62, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29038925

ABSTRACT

OBJECTIVES: To investigate whether miR-1260b can regulate migration and invasion of hepatocellular carcinoma (HCC) by targeting RGS22. RESULTS: miR-1260b was up-regulated in HCC tissues compared with their corresponding non-cancerous tissues. Over-expression of miR-1260b increased migration and invasion of HepG2 and SMMC-7721 cells associated with HCC. Regulator of G-protein signaling 22 (RGS22) was identified as a directly target of miR-1260b and was inhibited by miR-1260b. Knockdown of RGS22 increased proliferation of HCC cells. CONCLUSIONS: The new identified miR-1260b/RGS22 axis provides useful therapeutic methods for treatment of HCC deepening on our understanding of underlying mechanisms of HCC tumorigenesis.


Subject(s)
Antigens, Surface/metabolism , Carcinoma, Hepatocellular/pathology , Cell Movement , Cell Proliferation , GTP-Binding Protein Regulators/metabolism , Liver Neoplasms/pathology , MicroRNAs/metabolism , Carcinoma, Hepatocellular/physiopathology , Hep G2 Cells , Humans , Liver Neoplasms/physiopathology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...