Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 447
Filter
1.
Elife ; 122024 Apr 23.
Article in English | MEDLINE | ID: mdl-38651641

ABSTRACT

Inhibitory G alpha (GNAI or Gαi) proteins are critical for the polarized morphogenesis of sensory hair cells and for hearing. The extent and nature of their actual contributions remains unclear, however, as previous studies did not investigate all GNAI proteins and included non-physiological approaches. Pertussis toxin can downregulate functionally redundant GNAI1, GNAI2, GNAI3, and GNAO proteins, but may also induce unrelated defects. Here, we directly and systematically determine the role(s) of each individual GNAI protein in mouse auditory hair cells. GNAI2 and GNAI3 are similarly polarized at the hair cell apex with their binding partner G protein signaling modulator 2 (GPSM2), whereas GNAI1 and GNAO are not detected. In Gnai3 mutants, GNAI2 progressively fails to fully occupy the sub-cellular compartments where GNAI3 is missing. In contrast, GNAI3 can fully compensate for the loss of GNAI2 and is essential for hair bundle morphogenesis and auditory function. Simultaneous inactivation of Gnai2 and Gnai3 recapitulates for the first time two distinct types of defects only observed so far with pertussis toxin: (1) a delay or failure of the basal body to migrate off-center in prospective hair cells, and (2) a reversal in the orientation of some hair cell types. We conclude that GNAI proteins are critical for hair cells to break planar symmetry and to orient properly before GNAI2/3 regulate hair bundle morphogenesis with GPSM2.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go , Hair Cells, Auditory , Morphogenesis , Animals , Mice , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/physiology , Cell Polarity , GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein alpha Subunit, Gi2/genetics
2.
J Biol Chem ; 300(5): 107211, 2024 May.
Article in English | MEDLINE | ID: mdl-38522511

ABSTRACT

Highly homologous members of the Gαi family, Gαi1-3, have distinct tissue distributions and physiological functions, yet their biochemical and functional properties are very similar. We recently identified PDZ-RhoGEF (PRG) as a novel Gαi1 effector that is poorly activated by Gαi2. In a proteomic proximity labeling screen we observed a strong preference for Gαi1 relative to Gαi2 with respect to engagement of a broad range of potential targets. We investigated the mechanistic basis for this selectivity using PRG as a representative target. Substitution of either the helical domain (HD) from Gαi1 into Gαi2 or substitution of a single amino acid, A230 in Gαi2 with the corresponding D in Gαi1, largely rescues PRG activation and interactions with other potential Gαi targets. Molecular dynamics simulations combined with Bayesian network models revealed that in the GTP bound state, separation at the HD-Ras-like domain (RLD) interface is more pronounced in Gαi2 than Gαi1. Mutation of A230 to D in Gαi2 stabilizes HD-RLD interactions via ionic interactions with R145 in the HD which in turn modify the conformation of Switch III. These data support a model where D229 in Gαi1 interacts with R144 and stabilizes a network of interactions between HD and RLD to promote protein target recognition. The corresponding A230 in Gαi2 is unable to stabilize this network leading to an overall lower efficacy with respect to target interactions. This study reveals distinct mechanistic properties that could underly differential biological and physiological consequences of activation of Gαi1 or Gαi2 by G protein-coupled receptors.


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go , Signal Transduction , Humans , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/chemistry , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Molecular Dynamics Simulation , GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein alpha Subunit, Gi2/genetics , GTP-Binding Protein alpha Subunit, Gi2/chemistry , HEK293 Cells , Protein Domains , Protein Stability , Protein Binding
3.
Theranostics ; 14(5): 2190-2209, 2024.
Article in English | MEDLINE | ID: mdl-38505600

ABSTRACT

Here we explored the potential role of Gαi2 (G protein subunit alpha i2) in endothelial cell function and angiogenesis. Methods: Genetic methodologies such as shRNA, CRISPR/Cas9, dominant negative mutation, and overexpression were utilized to modify Gαi2 expression or regulate its function. Their effects on endothelial cell functions were assessed in vitro. In vivo, the endothelial-specific Gαi2 shRNA adeno-associated virus (AAV) was utilized to silence Gαi2 expression. The impact of this suppression on retinal angiogenesis in control mice and streptozotocin (STZ)-induced diabetic retinopathy (DR) mice was analyzed. Results: Analysis of single-cell RNA sequencing data revealed Gαi2 (GNAI2) was predominantly expressed in retinal endothelial cells and expression was increased in retinal endothelial cells following oxygen-induced retinopathy (OIR) in mice. Moreover, transcriptome analysis linking Gαi2 to angiogenesis-related processes/pathways, supported by increased Gαi2 expression in experimental OIR mouse retinas, highlighted its possible role in angiogenesis. In various endothelial cell types, shRNA-induced silencing and CRISPR/Cas9-mediated knockout (KO) of Gαi2 resulted in substantial reductions in cell proliferation, migration, invasion, and capillary tube formation. Conversely, Gαi2 over-expression in endothelial cells induced pro-angiogenic activities, enhancing cell proliferation, migration, invasion, and capillary tube formation. Furthermore, our investigation revealed a crucial role of Gαi2 in NFAT (nuclear factor of activated T cells) activation, as evidenced by the down-regulation of NFAT-luciferase reporter activity and pro-angiogenesis NFAT-targeted genes (Egr3, CXCR7, and RND1) in Gαi2-silenced or -KO HUVECs, which were up-regulated in Gαi2-overexpressing endothelial cells. Expression of a dominant negative Gαi2 mutation (S48C) also down-regulated NFAT-targeted genes, slowing proliferation, migration, invasion, and capillary tube formation in HUVECs. Importantly, in vivo experiments revealed that endothelial Gαi2 knockdown inhibited retinal angiogenesis in mice, with a concomitant down-regulation of NFAT-targeted genes in mouse retinal tissue. In contrast, Gαi2 over-expression in endothelial cells enhanced retinal angiogenesis in mice. Single-cell RNA sequencing data confirmed increased levels of Gαi2 specifically in retinal endothelial cells of mice with streptozotocin (STZ)-induced diabetic retinopathy (DR). Importantly, endothelial Gαi2 silencing ameliorated retinal pathological angiogenesis in DR mice. Conclusion: Our study highlights a critical role for Gαi2 in NFAT activation, endothelial cell activation and angiogenesis, offering valuable insights into potential therapeutic strategies for modulating these processes.


Subject(s)
Diabetic Retinopathy , Mice , Animals , Diabetic Retinopathy/drug therapy , GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein alpha Subunit, Gi2/pharmacology , Endothelial Cells/metabolism , Angiogenesis , Streptozocin/adverse effects , Oxygen/metabolism , RNA, Small Interfering/metabolism , Cell Proliferation
4.
Cell Death Dis ; 14(2): 143, 2023 02 20.
Article in English | MEDLINE | ID: mdl-36805440

ABSTRACT

Hepatocellular carcinoma (HCC) is a global health problem. Its incidence and mortality are increasing. Exploring novel therapeutic targets against HCC is important and urgent. We here explored the expression and potential function of Gαi2 (G protein subunit alpha i2) in HCC. The Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA-LIHC) database shows that the number of Gαi2 transcripts in HCC tissues is significantly higher than that in the normal liver tissues. Moreover, Gαi2 overexpression in HCC correlates with poor prognosis of the patients. Gαi2 mRNA and protein expression are also elevated in local HCC tissues and different human HCC cells. In patient-derived primary HCC cells and immortalized HepG2 cells, Gαi2 silencing (by targeted shRNA) or knockout (KO, by the dCas9-sgRNA method) largely suppressed cell proliferation and motility, while inducing cell cycle arrest and caspase-apoptosis activation. Moreover, Gαi2 silencing or KO-induced reactive oxygen species (ROS) production and oxidative injury in primary and HepG2 HCC cells. Whereas different antioxidants ameliorated Gαi2-shRNA-induced anti-HCC cell activity. Using a lentiviral construct, Gαi2 overexpression further augmented proliferation and motility of primary and immortalized HCC cells. Further studies revealed that the binding between the transcription factor early growth response zinc finger transcription factor 1 (EGR1) and Gαi2 DNA promoter was significantly increased in HCC tissues and cells. In vivo, intratumoral injection of Gαi2 shRNA adeno-associated virus significantly hindered HCC xenograft growth in nude mice. Moreover, the growth of Gαi2-KO HCC xenografts in the nude mice was remarkably slow. Gαi2 depletion, oxidative injury, and apoptosis induction were detected in Gαi2-silenced or Gαi2-KO HCC xenografts. Together, overexpressed Gαi2 is required for HCC cell growth in vitro and in vivo, representing as a novel and promising diagnosis marker and therapeutic target of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Mice , Humans , GTP-Binding Protein alpha Subunit, Gi2 , Carcinoma, Hepatocellular/genetics , Mice, Nude , Liver Neoplasms/genetics , Early Growth Response Protein 1
5.
Oxid Med Cell Longev ; 2022: 1254367, 2022.
Article in English | MEDLINE | ID: mdl-36275898

ABSTRACT

Purpose: Although the G protein subunit α i2 (GNAI2) is upregulated in multiple cancers, its prognostic value and exact role in the development of gastric cancer (GC) remain largely unknown. Methods: This study evaluated the effect of GNAI2 on the tumor microenvironment (TME) in GC, constructed an immune risk score (IRS) model based on differentially-expressed immune genes, and systematically correlated GNAI2 and epigenetic factor expression patterns with TME and IRS. Also, RT-qPCR, flow cytometry, Western blotting (WB), and transwell assays were carried out to explore the regulatory mechanism of GNAI2 in GC. Results: High GNAI2 expression was associated with poor prognosis. Cytokine activation, an increase in tumor-infiltrating immune cells (TIIC), and the accumulation of regulatory T cells in the tumor immune cycle were all promoted by the TME, which was significantly associated with GNAI2 expression. Two different differentially expressed mRNA (DER) modification patterns were determined. These two DERs-clusters had significantly different TME cell infiltrations and were classified as either noninflamed or immune-inflamed phenotypes. The IRS model constructed using differentially expressed genes (DEGs) had great potential in predicting GC prognosis. The IRS model was also used in assessing clinicopathological features, such as microsatellite instability (MSI) status, epithelial-mesenchymal transition (EMT) status, clinical stages, tumor mutational burden (TMB), and tumor immune dysfunction and exclusion (TIDE) scores. Low IRS scores were associated with high immune checkpoint gene expression. Cell and animal studies confirmed that GNAI2 activated PI3K/AKT pathway and promoted the growth and migration of GC cells. Conclusion: The IRS model can be used for survival prediction and GNAI2 serves as a candidate therapeutic target for GC patients.


Subject(s)
Stomach Neoplasms , Animals , Stomach Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Tumor Microenvironment/genetics , GTP-Binding Protein alpha Subunit, Gi2/genetics , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Gene Expression Regulation, Neoplastic , RNA, Messenger , Risk Factors , Cytokines/metabolism
6.
J Physiol Pharmacol ; 73(2)2022 Apr.
Article in English | MEDLINE | ID: mdl-36193969

ABSTRACT

The present study aimed to screen out the proteins with significantly differential expression through the proteomics study of Tianxiangdan intervention in rats with myocardial ischemia as well as elucidate the mechanism of the intervention. In total, 54 Wistar rats (male, 6-8 weeks old) were randomly divided into the blank group, sham operation group, and model group, with 6 rats in each, together with the model + low dose group, model + medium-dose group, and model + high dose group, with 12 rats in each. Upon successful construction of the ischemic model, low, medium, and high doses, respectively, of Tianxiangdan were administered in the groups. The rat model of coronary heart disease (CHD) with myocardial ischemia was prepared by ligating the coronary artery. The tandem mass tag-labeled quantitative proteomics technology was adopted to observe the differentially expressed proteins in the myocardium of the model rats under the action of Tianxiangdan to find the target proteins for the treatment of myocardial ischemia in CHD. A total of 3122 proteins were identified. Combined with the references, tropomyosin alpha-3 chain (TPM3), protein kinase C delta (PRKCD), myosin heavy chain 10 (MYH10), MYH6, G protein subunit alpha i2 (GNAI2), and other proteins were screened out. Western blotting was adopted for the proteomics validation, and it was found that compared with the sham operation group, the expression levels of the GNAI2, TPM3, and MYH10 proteins were upregulated in the myocardial ischemia model group but downregulated after the administration of Tianxiangdan; the differences were statistically significant (p<0.05). We conclude that Tianxiangdan could improve myocardial ischemia by downregulating the proteins, including GNAI2, TPM3, and MYH10, which might be potential targets of Tianxiangdan in the treatment of myocardial infarction.


Subject(s)
Myocardial Ischemia , Tropomyosin , Animals , Male , Rats , GTP-Binding Protein alpha Subunit, Gi2 , Myocardial Ischemia/drug therapy , Myocardium , Myosin Heavy Chains , Protein Kinase C-delta , Proteomics , Rats, Wistar
7.
Int J Biol Sci ; 18(5): 2202-2219, 2022.
Article in English | MEDLINE | ID: mdl-35342351

ABSTRACT

Background: Intervertebral disc degeneration (IDD), the main cause of low back pain, is closely related to the inflammatory microenvironment in the nucleus pulposus (NP). Tumor necrosis factor-α (TNF-α) plays an important role in inflammation-related metabolic disturbance of NP cells. Melatonin has been proven to regulate the metabolism of NP cells, but whether it can protect NP cells from TNF-α-induced damage is still unclear. Therefore, this study aims to investigate the role and specific mechanism of melatonin on regulating the metabolism of NP cells in the inflammatory microenvironment. Methods: Western blotting, RT-qPCR and immunohistochemistry were used to detect the expression of melatonin membrane receptors (MTNR1A/B) and TNF-α in human NP tissues. In vitro, human primary NP cells were treated with or without vehicle, TNF-α and melatonin. And the metabolic markers were also detected by western blotting and RT-qPCR. The activity of NF-κB signaling and Hippo/YAP signaling were assessed by western blotting and immunofluorescence. Membrane receptors inhibitors, pathway inhibitors, lentiviral infection, plasmids transfection and immunoprecipitation were used to explore the specific mechanism of melatonin. In vivo, the rat IDD model was constructed and melatonin was injected intraperitoneally to evaluate its therapeutical effect on IDD. Results: The upregulation of TNF-α and downregulation of melatonin membrane receptors (MTNR1A/B) were observed in degenerative NP tissues. Then we demonstrated that melatonin could alleviate the development of IDD in a rat model and reverse TNF-α-impaired metabolism of NP cells in vitro. Further investigation revealed that the protective effects of melatonin on NP cells mainly rely on MTNR1B, which subsequently activates Gαi2 protein. The activation of Gαi2 could upregulate the yes-associated protein (YAP) level, resulting in anabolic enhancement of NP cells. In addition, melatonin-mediated YAP upregulation increased the expression of IκBα and suppressed the TNF-α-induced activation of the NF-κB pathway, thereby inhibiting the catabolism of NP cells. Conclusions: Our results revealed that melatonin can reverse TNF-α-impaired metabolism of NP cells via the MTNR1B/Gαi2/YAP axis and suggested that melatonin can be used as a potential therapeutic drug in the treatment of IDD.


Subject(s)
Intervertebral Disc Degeneration , Melatonin , Nucleus Pulposus , Animals , GTP-Binding Protein alpha Subunit, Gi2/metabolism , GTP-Binding Protein alpha Subunit, Gi2/pharmacology , Humans , Intervertebral Disc Degeneration/metabolism , Melatonin/metabolism , Melatonin/pharmacology , Melatonin/therapeutic use , NF-kappa B/metabolism , Nucleus Pulposus/metabolism , Rats , Receptor, Melatonin, MT2/metabolism , Tumor Necrosis Factor-alpha/metabolism
8.
Nat Commun ; 13(1): 674, 2022 02 03.
Article in English | MEDLINE | ID: mdl-35115535

ABSTRACT

Conductin/axin2 is a scaffold protein negatively regulating the pro-proliferative Wnt/ß-catenin signaling pathway. Accumulation of scaffold proteins in condensates frequently increases their activity, but whether condensation contributes to Wnt pathway inhibition by conductin remains unclear. Here, we show that the Gαi2 subunit of trimeric G-proteins induces conductin condensation by targeting a polymerization-inhibiting aggregon in its RGS domain, thereby promoting conductin-mediated ß-catenin degradation. Consistently, transient Gαi2 expression inhibited, whereas knockdown activated Wnt signaling via conductin. Colorectal cancers appear to evade Gαi2-induced Wnt pathway suppression by decreased Gαi2 expression and inactivating mutations, associated with shorter patient survival. Notably, the Gαi2-activating drug guanabenz inhibited Wnt signaling via conductin, consequently reducing colorectal cancer growth in vitro and in mouse models. In summary, we demonstrate Wnt pathway inhibition via Gαi2-triggered conductin condensation, suggesting a tumor suppressor function for Gαi2 in colorectal cancer, and pointing to the FDA-approved drug guanabenz for targeted cancer therapy.


Subject(s)
Axin Protein/genetics , Colorectal Neoplasms/genetics , GTP-Binding Protein alpha Subunit, Gi2/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics , Adrenergic alpha-2 Receptor Agonists/pharmacology , Animals , Axin Protein/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Gene Expression Regulation, Neoplastic , Guanabenz/pharmacology , HEK293 Cells , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mutation , Wnt Signaling Pathway/drug effects , Xenograft Model Antitumor Assays/methods , beta Catenin/metabolism
9.
Sci Signal ; 15(717): eabi9869, 2022 01 18.
Article in English | MEDLINE | ID: mdl-35041463

ABSTRACT

G protein­coupled receptors (GPCRs) that couple to the Gαi family of G proteins are key regulators of cell and tissue physiology. Our previous work has revealed new roles for Gαi in regulating the migration of neutrophils and fibrosarcoma cells downstream of activated chemoattractant receptors. Here, we used an intact cell proximity­based labeling coupled to tandem mass tag (TMT)­based quantitative proteomics analysis to identify proteins that selectively interacted with the GTP-bound form of Gαi1. Multiple targets were identified and validated with a BioID2-tagged, constitutively active Gαi1 mutant, suggesting a network of interactions for activated GαI proteins in intact cells. We showed that active Gαi1, but not Gαi2, stimulated one candidate protein, PDZ-RhoGEF (PRG), despite more than 85% sequence identity between the G proteins. We also demonstrated in primary human neutrophils that active Gαi likely regulated the polarization of phosphorylated myosin light chain, a process critical for migration, through the activation of PRG. The identification and characterization of new targets directly or indirectly regulated by Gαi will aid in the investigation of the functional roles of Gαi-coupled GPCRs in multiple biological processes.


Subject(s)
Biological Phenomena , Proteomics , GTP-Binding Protein alpha Subunit, Gi2 , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction/physiology
10.
Nature ; 597(7877): 571-576, 2021 09.
Article in English | MEDLINE | ID: mdl-34497422

ABSTRACT

The adenosine A1 receptor (A1R) is a promising therapeutic target for non-opioid analgesic agents to treat neuropathic pain1,2. However, development of analgesic orthosteric A1R agonists has failed because of a lack of sufficient on-target selectivity as well as off-tissue adverse effects3. Here we show that [2-amino-4-(3,5-bis(trifluoromethyl)phenyl)thiophen-3-yl)(4-chlorophenyl)methanone] (MIPS521), a positive allosteric modulator of the A1R, exhibits analgesic efficacy in rats in vivo through modulation of the increased levels of endogenous adenosine that occur in the spinal cord of rats with neuropathic pain. We also report the structure of the A1R co-bound to adenosine, MIPS521 and a Gi2 heterotrimer, revealing an extrahelical lipid-detergent-facing allosteric binding pocket that involves transmembrane helixes 1, 6 and 7. Molecular dynamics simulations and ligand kinetic binding experiments support a mechanism whereby MIPS521 stabilizes the adenosine-receptor-G protein complex. This study provides proof of concept for structure-based allosteric drug design of non-opioid analgesic agents that are specific to disease contexts.


Subject(s)
Analgesia , Receptor, Adenosine A1/metabolism , Adenosine/chemistry , Adenosine/metabolism , Allosteric Regulation/drug effects , Analgesia/methods , Animals , Binding Sites , Disease Models, Animal , Female , GTP-Binding Protein alpha Subunit, Gi2/chemistry , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Hyperalgesia/drug therapy , Lipids , Male , Neuralgia/drug therapy , Neuralgia/metabolism , Protein Stability/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A1/chemistry , Signal Transduction/drug effects
11.
Nat Commun ; 12(1): 5043, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34413292

ABSTRACT

Skeletal muscle has a remarkable ability to regenerate owing to its resident stem cells (also called satellite cells, SCs). SCs are normally quiescent; when stimulated by damage, they activate and expand to form new fibers. The mechanisms underlying SC proliferative progression remain poorly understood. Here we show that DHX36, a helicase that unwinds RNA G-quadruplex (rG4) structures, is essential for muscle regeneration by regulating SC expansion. DHX36 (initially named RHAU) is barely expressed at quiescence but is highly induced during SC activation and proliferation. Inducible deletion of Dhx36 in adult SCs causes defective proliferation and muscle regeneration after damage. System-wide mapping in proliferating SCs reveals DHX36 binding predominantly to rG4 structures at various regions of mRNAs, while integrated polysome profiling shows that DHX36 promotes mRNA translation via 5'-untranslated region (UTR) rG4 binding. Furthermore, we demonstrate that DHX36 specifically regulates the translation of Gnai2 mRNA by unwinding its 5' UTR rG4 structures and identify GNAI2 as a downstream effector of DHX36 for SC expansion. Altogether, our findings uncover DHX36 as an indispensable post-transcriptional regulator of SC function and muscle regeneration acting through binding and unwinding rG4 structures at 5' UTR of target mRNAs.


Subject(s)
5' Untranslated Regions , DEAD-box RNA Helicases/metabolism , G-Quadruplexes , Muscles/cytology , Regeneration/physiology , Stem Cells/cytology , Animals , Animals, Genetically Modified , Cells, Cultured , Disease Models, Animal , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Gene Expression Regulation , Humans , Mice , Muscles/metabolism , Myoblasts/metabolism , Polyribosomes/metabolism , Protein Biosynthesis , RNA, Messenger/genetics , Stem Cells/metabolism
12.
Biomolecules ; 11(8)2021 08 14.
Article in English | MEDLINE | ID: mdl-34439877

ABSTRACT

Increased expression of GNAi2, which encodes the α-subunit of G-protein i2, has been correlated with the late-stage progression of ovarian cancer. GNAi2, also referred to as the proto-oncogene gip2, transduces signals from lysophosphatidic acid (LPA)-activated LPA-receptors to oncogenic cellular responses in ovarian cancer cells. To identify the oncogenic program activated by gip2, we carried out micro-array-based transcriptomic and bioinformatic analyses using the ovarian cancer cell-line SKOV3, in which the expression of GNAi2/gip2 was silenced by specific shRNA. A cut-off value of 5-fold change in gene expression (p < 0.05) indicated that a total of 264 genes were dependent upon gip2-expression with 136 genes coding for functional proteins. Functional annotation of the transcriptome indicated the hitherto unknown role of gip2 in stimulating the expression of oncogenic/growth-promoting genes such as KDR/VEGFR2, CCL20, and VIP. The array results were further validated in a panel of High-Grade Serous Ovarian Carcinoma (HGSOC) cell lines that included Kuramochi, OVCAR3, and OVCAR8 cells. Gene set enrichment analyses using DAVID, STRING, and Cytoscape applications indicated the potential role of the gip2-stimulated transcriptomic network involved in the upregulation of cell proliferation, adhesion, migration, cellular metabolism, and therapy resistance. The results unravel a multi-modular network in which the hub and bottleneck nodes are defined by ACKR3/CXCR7, IL6, VEGFA, CYCS, COX5B, UQCRC1, UQCRFS1, and FYN. The identification of these genes as the critical nodes in GNAi2/gip2 orchestrated onco-transcriptome establishes their role in ovarian cancer pathophysiology. In addition, these results also point to these nodes as potential targets for novel therapeutic strategies.


Subject(s)
Carcinoma, Ovarian Epithelial/metabolism , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Gene Expression Regulation, Neoplastic , Ovarian Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , Proto-Oncogene Mas , Transcriptome
13.
Genes (Basel) ; 12(8)2021 07 25.
Article in English | MEDLINE | ID: mdl-34440304

ABSTRACT

GNAI2 (G protein subunit alpha i2) is a signaling modulator or transducer, involved in several transmembrane signaling systems, that plays a vital role in the melanogenesis signaling pathway. However, whether GNAI2 regulates cell proliferation and apoptosis in rabbit melanocytes is not known. We found that GNAI2 was differentially expressed in rabbits with different coat colors using qRT-PCR and Wes assays. Furthermore, it was observed that the rabbits with black skin had the highest GNAI2 levels, and those with white skin had the lowest expression. The coding sequence of GNAI2 was successfully cloned and inserted into pcDNA3.1 and pcDNA3.1-Myc vectors. It was observed that the GNAI2 protein was mainly localized in the cytoplasm using the indirect immunofluorescence staining assay. Overexpression of GNAI2 significantly increased melanin content, promoted melanocyte proliferation, and inhibited melanocyte apoptosis. On the contrary, the knockdown of GNAI2 using siRNA had the opposite effect. In addition, GNAI2 significantly increased the mRNA expression levels of the melanin-related genes TYR, GPNMB, PMEL, and DCT in rabbit melanocytes. The results suggested that GNAI2 regulated melanocyte development by promoting melanocyte proliferation and inhibiting apoptosis.


Subject(s)
Apoptosis/physiology , Cell Proliferation/physiology , GTP-Binding Protein alpha Subunit, Gi2/physiology , Melanocytes/cytology , Animals , Cytoplasm/metabolism , GTP-Binding Protein alpha Subunit, Gi2/genetics , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Gene Knockdown Techniques , Hair Color , Melanins/biosynthesis , Melanocytes/metabolism , Rabbits
14.
Nat Commun ; 12(1): 4452, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34294713

ABSTRACT

Atherosclerosis-associated cardiovascular disease is one of the main causes of death and disability among patients with diabetes mellitus. However, little is known about the impact of S-nitrosylation in diabetes-accelerated atherosclerosis. Here, we show increased levels of S-nitrosylation of guanine nucleotide-binding protein G(i) subunit alpha-2 (SNO-GNAI2) at Cysteine 66 in coronary artery samples from diabetic patients with atherosclerosis, consistently with results from mice. Mechanistically, SNO-GNAI2 acted by coupling with CXCR5 to dephosphorylate the Hippo pathway kinase LATS1, thereby leading to nuclear translocation of YAP and promoting an inflammatory response in endothelial cells. Furthermore, Cys-mutant GNAI2 refractory to S-nitrosylation abrogated GNAI2-CXCR5 coupling, alleviated atherosclerosis in diabetic mice, restored Hippo activity, and reduced endothelial inflammation. In addition, we showed that melatonin treatment restored endothelial function and protected against diabetes-accelerated atherosclerosis by preventing GNAI2 S-nitrosylation. In conclusion, SNO-GNAI2 drives diabetes-accelerated atherosclerosis by coupling with CXCR5 and activating YAP-dependent endothelial inflammation, and reducing SNO-GNAI2 is an efficient strategy for alleviating diabetes-accelerated atherosclerosis.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/metabolism , Diabetic Angiopathies/etiology , Diabetic Angiopathies/metabolism , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cells, Cultured , Cysteine/chemistry , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , GTP-Binding Protein alpha Subunit, Gi2/chemistry , GTP-Binding Protein alpha Subunit, Gi2/genetics , Hippo Signaling Pathway , Human Umbilical Vein Endothelial Cells , Humans , Male , Melatonin/pharmacology , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Nitric Oxide Synthase Type II/metabolism , Nitroso Compounds/chemistry , Nitroso Compounds/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, CXCR5/deficiency , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism , Transcription Factors/metabolism , YAP-Signaling Proteins
15.
Hepatology ; 74(6): 3110-3126, 2021 12.
Article in English | MEDLINE | ID: mdl-34322898

ABSTRACT

BACKGROUND AND AIMS: NASH is an advanced stage of liver disease accompanied by lipid accumulation, inflammation, and liver fibrosis. Guanine nucleotide-binding protein G(i) subunit alpha-2 (GNAI2) is a member of the "inhibitory" class of α-subunits, and recent studies showed that Gnai2 deficiency is known to cause reduced weight in mice. However, the role of GNAI2 in hepatocytes, particularly in the context of liver inflammation and lipid metabolism, remains to be elucidated. Herein, we aim to ascertain the function of GNAI2 in hepatocytes and its impact on the development of NASH. APPROACH AND RESULTS: Human liver tissues were obtained from NASH patients and healthy persons to evaluate the expression and clinical relevance of GNAI2. In addition, hepatocyte-specific Gnai2-deficient mice (Gnai2hep-/- ) were fed either a Western diet supplemented with fructose in drinking water (WDF) for 16 weeks or a methionine/choline-deficient diet (MCD) for 6 weeks to investigate the regulatory role and underlying mechanism of Gnai2 in NASH. GNAI2 was significantly up-regulated in liver tissues of patients with NASH. Following feeding with WDF or MCD diets, livers from Gnai2hep-/- mice had reduced steatohepatitis with suppression of markers of inflammation and an increase in lipophagy compared to Gnai2flox/flox mice. Toll-like receptor 4 signals through nuclear factor kappa B to trigger p65-dependent transcription of Gnai2. Intriguingly, immunoprecipitation, immunofluorescence, and mass spectrometry identified peroxiredoxin 1 (PRDX1) as a binding partner of GNAI2. Moreover, the function of PRDX1 in the suppression of TNF receptor-associated factor 6 ubiquitin-ligase activity and glycerophosphodiester phosphodiesterase domain-containing 5-related phosphatidylcholine metabolism was inhibited by GNAI2. Suppression of GNAI2 combined with overexpression of PRDX1 reversed the development of steatosis and fibrosis in vivo. CONCLUSIONS: GNAI2 is a major regulator that leads to the development of NASH. Thus, inhibition of GNAI2 could be an effective therapeutic target for the treatment of NASH.


Subject(s)
GTP-Binding Protein alpha Subunit, Gi2/metabolism , Non-alcoholic Fatty Liver Disease/immunology , Peroxiredoxins/metabolism , Adult , Animals , Autophagy/immunology , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , GTP-Binding Protein alpha Subunit, Gi2/genetics , Hepatocytes , Humans , Liver/immunology , Liver/pathology , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Protein Binding/immunology , Signal Transduction/immunology , Young Adult
16.
Front Immunol ; 12: 679856, 2021.
Article in English | MEDLINE | ID: mdl-34135907

ABSTRACT

Neutrophil trafficking, homeostatic and pathogen elicited, depends upon chemoattractant receptors triggering heterotrimeric G-protein Gαißγ signaling, whose magnitude and kinetics are governed by RGS protein/Gαi interactions. RGS proteins typically limit Gαi signaling by reducing the duration that Gαi subunits remain GTP bound and able to activate downstream effectors. Yet how in totality RGS proteins shape neutrophil chemoattractant receptor activated responses remains unclear. Here, we show that C57Bl/6 mouse neutrophils containing a genomic knock-in of a mutation that disables all RGS protein-Gαi2 interactions (G184S) cannot properly balance chemoattractant receptor signaling, nor appropriately respond to inflammatory insults. Mutant neutrophils accumulate in mouse bone marrow, spleen, lung, and liver; despite neutropenia and an intrinsic inability to properly mobilize from the bone marrow. In vitro they rapidly adhere to ICAM-1 coated plates, but in vivo they poorly adhere to blood vessel endothelium. Those few neutrophils that cross blood vessels and enter tissues migrate haphazardly. Following Concanavalin-A administration fragmented G184S neutrophils accumulate in liver sinusoids leading to thrombo-inflammation and perivasculitis. Thus, neutrophil Gαi2/RGS protein interactions both limit and facilitate Gαi2 signaling thereby promoting normal neutrophil trafficking, aging, and clearance.


Subject(s)
Cellular Senescence , Chemotaxis, Leukocyte , GTP-Binding Protein alpha Subunit, Gi2/genetics , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Signal Transduction , Animals , Bone Marrow Transplantation , Cellular Senescence/genetics , Cellular Senescence/immunology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/genetics , Chemotaxis, Leukocyte/immunology , Humans , Immunophenotyping , Male , Mice , Neutropenia/etiology , Neutrophils/drug effects , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism , Receptors, Interleukin-8B/antagonists & inhibitors , Receptors, Interleukin-8B/metabolism
17.
Mol Pharmacol ; 100(3): 217-223, 2021 09.
Article in English | MEDLINE | ID: mdl-34135098

ABSTRACT

Regulators of G protein signaling (RGS) proteins modulate signaling by G protein-coupled receptors. Using a knock-in transgenic mouse model with a mutation in Gαo that does not bind RGS proteins (RGS-insensitive), we determined the effect of RGS proteins on presynaptic µ opioid receptor (MOR)-mediated inhibition of GABA release in the ventrolateral periaqueductal gray (vlPAG). The MOR agonists [d-Ala2, N-MePhe4, Gly-ol]-enkephalin (DAMGO) and met-enkephalin (ME) inhibited evoked inhibitory postsynaptic currents (eIPSCs) in the RGS-insensitive mice compared with wild-type (WT) littermates, respectively. Fentanyl inhibited eIPSCs similarly in both WT and RGS-insensitive mice. There were no differences in opioid agonist inhibition of spontaneous GABA release between the genotypes. To further probe the mechanism underlying these differences between opioid inhibition of evoked and spontaneous GABA release, specific myristoylated Gα peptide inhibitors for Gαo1 and Gαi1-3 that block receptor-G protein interactions were used to test the preference of agonists for MOR-Gα complexes. The Gαo1 inhibitor reduced DAMGO inhibition of eIPSCs, but Gαi1-3 inhibitors had no effect. Both Gαo1 and Gαi1-3 inhibitors separately reduced fentanyl inhibition of eIPSCs but had no effects on ME inhibition. Gαi1-3 inhibitors blocked the inhibitory effects of ME and fentanyl on miniature postsynaptic current (mIPSC) frequency, but both Gαo1 and Gαi1-3 inhibitors were needed to block the effects of DAMGO. Finally, baclofen-mediated inhibition of GABA release is unaffected in the RGS-insensitive mice and in the presence of Gαo1 and Gαi1-3 inhibitor peptides, suggesting that GABAB receptor coupling to G proteins in vlPAG presynaptic terminals is different than MOR coupling. SIGNIFICANCE STATEMENT: Presynaptic µ opioid receptors (MORs) in the ventrolateral periaqueductal gray are critical for opioid analgesia and are negatively regulated by RGS proteins. These data in RGS-insensitive mice provide evidence that MOR agonists differ in preference for Gαo versus Gαi and regulation by RGS proteins in presynaptic terminals, providing a mechanism for functional selectivity between agonists. The results further define important differences in MOR and GABAB receptor coupling to G proteins that could be exploited for new pain therapies.


Subject(s)
GTP-Binding Protein alpha Subunit, Gi2/physiology , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Presynaptic Terminals/physiology , Receptors, Opioid, mu/physiology , gamma-Aminobutyric Acid/metabolism , Analgesics, Opioid/pharmacology , Animals , Baclofen/pharmacology , Female , GTP-Binding Protein alpha Subunit, Gi2/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gi-Go/genetics , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Mice , Mice, Transgenic , Models, Animal , RGS Proteins/metabolism , Receptors, GABA-B/metabolism , Receptors, Opioid, mu/agonists
18.
Neuropsychopharmacology ; 46(5): 920-927, 2021 04.
Article in English | MEDLINE | ID: mdl-33531622

ABSTRACT

The mu opioid receptor (MOR) is a G protein-coupled receptor that plays an essential role in reward and hedonic processes, and that has been implicated in disorders such as depression and addiction. Over the last decade, several brain imaging studies in depressed patients have consistently found that dysregulation of MOR function occurs in particular in the anterior insular cortex, an important brain site for the perception of internal states and emotional regulation. To investigate molecular mechanisms that may underlie these effects, here we assessed genetic polymorphisms, expression, and functional G-protein coupling of MOR in a large post-mortem cohort (N = 95) composed of depressed individuals who died by suicide, and healthy controls. Results indicated that depression, but not comorbid substance use disorder or acute opiate consumption, was associated with increased MOR activity. This effect was partly explained by a specific increase in expression of the inhibitory alpha G-protein subunit GNAI2. Consistent with previous neuroimaging studies, our findings support the notion that enhanced endogenous opioidergic tone in the anterior insula may buffer negative affective states in depressed individuals, a mechanism that could potentially contribute to the antidepressant efficacy of emerging opioid-based medications.


Subject(s)
Brain , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Receptors, Opioid, mu , Analgesics, Opioid , Brain/metabolism , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/metabolism , Emotions , Humans , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism
19.
Pharmacol Rep ; 73(4): 1122-1135, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33641090

ABSTRACT

BACKGROUND: Alpha2A-adrenergic (α2A-AR) and cannabinoid CB1 (CB1-R) receptors exert their functions modulating multiple signaling pathways, including MEK-ERK (extracellular signal-regulated kinases) and FADD (Fas-associated protein with death domain) cascades. These molecules are relevant in finding biased agonists with fewer side effects, but the mechanisms involving their modulations by α2A-AR- and CB1-R in vivo are unclear. This study investigated the roles of Gαi2 and Gαz proteins in mediating α2A-AR- and CB1-R-induced alterations of MEK-ERK and FADD phosphorylation (p-) in mouse brain cortex. METHODS: Gαi2 or Gαz protein knockdown was induced in mice with selective antisense oligodeoxinucleotides (ODNs; 3 nmol/day, 5 days) prior to UK-14,304 (UK or brimonidine; 1 mg/kg) or WIN55212-2 (WIN; 8 mg/kg) acute treatments. Inactivated (p-T286) MEK1, activated (p-S217/221) MEK1/2, activated (p-T202/Y204) ERK1/2, p-S191 FADD, and the corresponding total forms of these proteins were quantified by immunoblotting. RESULTS: Increased (+ 88%) p-T286 MEK1 cortical density, with a concomitant reduction (-43%) of activated ERK was observed in UK-treated mice. Both effects were attenuated by Gαi2 or Gαz antisense ODNs. Contrastingly, WIN induced Gαi2- and Gαz-independent upregulations of p-T286 MEK1 (+ 63%), p-S217/221 MEK1/2 (+ 86%), and activated ERK (+ 111%) in brain. Pro-apoptotic FADD was downregulated (- 34 to 39%) following UK and WIN administration, whereas the neuroprotective p-S191 FADD was increased (+ 74%) in WIN-treated mice only. None of these latter effects required from Gαi2 or Gαz protein integrity. CONCLUSION: The results indicate that α2A-AR (UK), but not CB1-R (WIN), agonists use Gαi2 and Gαz proteins to modulate MEK-ERK, but not FADD, pathway in mouse brain cortex.


Subject(s)
Cerebral Cortex/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fas-Associated Death Domain Protein/metabolism , GTP-Binding Protein alpha Subunit, Gi2/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Receptor, Cannabinoid, CB1/metabolism , Signal Transduction/physiology , Animals , Down-Regulation/physiology , Male , Mice , Phosphorylation/physiology , Up-Regulation/physiology
20.
Mol Biol Rep ; 48(1): 435-449, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33386590

ABSTRACT

Plant microRNAs have shown the capacity to regulate mammalian systems. The potential bioactivity of miR171vr, an isoform of the plant miR171, on human embryonic kidney 293 (HEK293) cells was investigated. Bioinformatics simulations revealed that human G protein subunit alpha 12 (GNA12) transcript could represent an excellent target for miR171vr. To confirm this prediction, in vitro experiments were performed using a synthetic microRNA designed on miR171vr sequence. MiR-treated cells showed a significant decrease of GNA12 mRNA and protein levels, confirming the putative cross-kingdom interaction. In addition, miR171vr determined the modulation of GNA12 downstream signaling factors, including mTOR, as expected. Finally, the effect of the plant miRNA on HEK293 cell growth and its stability in presence of several stressors, such as those miming digestive processes and procedures for preparing food, were evaluated. All this preliminary evidence would suggest that miR171vr, introduced by diet or as supplement in gene therapies, could potentially influence human gene expression, especially for treating disorders where GNA12 is over-expressed (i.e. oral cancer, breast and prostate adenocarcinoma) or mTOR kinase is down-regulated (e.g. obesity, type 2 diabetes, neurodegeneration).


Subject(s)
GTP-Binding Protein alpha Subunit, Gi2/genetics , MicroRNAs/genetics , Protein Interaction Maps/genetics , TOR Serine-Threonine Kinases/genetics , Cell Proliferation/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/therapy , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , MicroRNAs/pharmacology , Neoplasms/genetics , Neoplasms/therapy , Obesity/genetics , Obesity/therapy , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...