Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 481
Filtrar
1.
J Biol Chem ; 300(4): 107127, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38432633

RESUMEN

Regulators of G protein signaling (RGS) proteins constrain G protein-coupled receptor (GPCR)-mediated and other responses throughout the body primarily, but not exclusively, through their GTPase-activating protein activity. Asthma is a highly prevalent condition characterized by airway hyper-responsiveness (AHR) to environmental stimuli resulting in part from amplified GPCR-mediated airway smooth muscle contraction. Rgs2 or Rgs5 gene deletion in mice enhances AHR and airway smooth muscle contraction, whereas RGS4 KO mice unexpectedly have decreased AHR because of increased production of the bronchodilator prostaglandin E2 (PGE2) by lung epithelial cells. Here, we found that knockin mice harboring Rgs4 alleles encoding a point mutation (N128A) that sharply curtails RGS4 GTPase-activating protein activity had increased AHR, reduced airway PGE2 levels, and augmented GPCR-induced bronchoconstriction compared with either RGS4 KO mice or WT controls. RGS4 interacted with the p85α subunit of PI3K and inhibited PI3K-dependent PGE2 secretion elicited by transforming growth factor beta in airway epithelial cells. Together, these findings suggest that RGS4 affects asthma severity in part by regulating the airway inflammatory milieu in a G protein-independent manner.


Asunto(s)
Asma , Proteínas RGS , Animales , Humanos , Ratones , Asma/metabolismo , Asma/genética , Asma/patología , Broncoconstricción/genética , Dinoprostona/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/patología , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/metabolismo , Hipersensibilidad Respiratoria/metabolismo , Hipersensibilidad Respiratoria/genética , Hipersensibilidad Respiratoria/patología , Proteínas RGS/metabolismo , Proteínas RGS/genética , Línea Celular
2.
Cell Mol Biol Lett ; 28(1): 102, 2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38066447

RESUMEN

BACKGROUND: Acute respiratory distress syndrome (ARDS) is a disease with high mortality and morbidity. Regulator of G protein signaling protein 6 (RGS6), identified as a tumor suppressor gene, has received increasing attention owing to its close relationship with oxidative stress and inflammation. However, the association between ARDS and RGS6 has not been reported. METHODS: Congruously regulated G protein-coupled receptor (GPCR)-related genes and differentially expressed genes (DEGs) in an acute lung injury (ALI) model were identified, and functional enrichment analysis was conducted. In an in vivo study, the effects of RGS6 knockout were studied in a mouse model of ALI induced by lipopolysaccharide (LPS). HE staining, ELISA, and immunohistochemistry were used to evaluate pathological changes and the degree of inflammation. In vitro, qRT‒PCR, immunofluorescence staining, and western blotting were used to determine the dynamic changes in RGS6 expression in cells. The RGS6 overexpression plasmid was constructed for transfection. qRT‒PCR was used to assess proinflammatory factors transcription. Western blotting and flow cytometry were used to evaluate apoptosis and reactive oxygen species (ROS) production. Organoid culture was used to assess the stemness and self-renewal capacity of alveolar epithelial type II cells (AEC2s). RESULTS: A total of 110 congruously regulated genes (61 congruously upregulated and 49 congruously downregulated genes) were identified among GPCR-related genes and DEGs in the ALI model. RGS6 was downregulated in vivo and in vitro in the ALI model. RGS6 was expressed in the cytoplasm and accumulated in the nucleus after LPS stimulation. Compared with the control group, we found higher mortality, more pronounced body weight changes, more serious pulmonary edema and pathological damage, and more neutrophil infiltration in the RGS6 knockout group upon LPS stimulation in vivo. Moreover, AEC2s loss was significantly increased upon RGS6 knockout. Organoid culture assays showed slower alveolar organoid formation, fewer alveolar organoids, and impaired development of new structures after passaging upon RGS6 knockout. In addition, RGS6 overexpression decreased ROS production as well as proinflammatory factor transcription in macrophages and decreased apoptosis in epithelial cells. CONCLUSIONS: RGS6 plays a protective role in ALI not only in early inflammatory responses but also in endogenous lung stem cell regeneration.


Asunto(s)
Lesión Pulmonar Aguda , Proteínas RGS , Síndrome de Dificultad Respiratoria , Animales , Ratones , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/metabolismo , Proteínas de Unión al GTP/efectos adversos , Proteínas de Unión al GTP/metabolismo , Inflamación/patología , Lipopolisacáridos , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno/metabolismo , Síndrome de Dificultad Respiratoria/genética , Síndrome de Dificultad Respiratoria/metabolismo , Proteínas RGS/metabolismo
3.
Biol Direct ; 18(1): 78, 2023 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-37986113

RESUMEN

BACKGROUND: Regulator of G protein signaling 5 (RGS5), as a negative regulator of G protein-coupled receptor (GPCR) signaling, is highly expressed in arterial VSMCs and pericytes, which is involved in VSMC phenotypic heterogeneity and vascular remodeling in tumors. However, its role in normal and tumor vascular remodeling is controversial. METHODS: RGS5 knockout (Rgs5-KO) mice and RGS5 overexpression or knockdown in VSMCs in vivo by adeno-associated virus type 9 (AAV) carrying RGS5 cDNA or small hairpin RNA (shRNA) targeting RGS5 were used to determine the functional significance of RGS5 in vascular inflammation. RGS5 expression in the triple-negative (TNBCs) and non-triple-negative breast cancers (Non-TNBCs) was determined by immunofluorescent and immunohistochemical staining. The effect of breast cancer cell-conditioned media (BC-CM) on the pro-inflammatory phenotype of VSMCs was measured by phagocytic activity assays, adhesion assay and Western blot. RESULTS: We identified that knockout and VSMC-specific knockdown of RGS5 exacerbated accumulation and pyroptosis of pro-inflammatory VSMCs, resulting in vascular remodeling, which was negated by VSMC-specific RGS5 overexpression. In contrast, in the context of breast cancer tissues, the role of RGS5 was completely disrupted. RGS5 expression was increased in the triple-negative breast cancer (TNBC) tissues and in the tumor blood vessels, accompanied with an extensive vascular network. VSMCs treated with BC-CM displayed enhanced pro-inflammatory phenotype and higher adherent with macrophages. Furthermore, tumor-derived RGS5 could be transferred into VSMCs. CONCLUSIONS: These findings suggest that tumor microenvironment shifts the function of RGS5 from anti-inflammation to pro-inflammation and induces the pro-inflammatory phenotype of VSMCs that is favorable for tumor metastasis.


Asunto(s)
Neoplasias , Proteínas RGS , Ratones , Animales , Proteínas RGS/genética , Proteínas RGS/metabolismo , Remodelación Vascular/genética , Músculo Liso Vascular/metabolismo , Microambiente Tumoral , Ratones Noqueados , Homeostasis , Inflamación , Proliferación Celular
4.
Cell Commun Signal ; 21(1): 316, 2023 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-37924113

RESUMEN

G protein-coupled receptors (GPCRs) play a key role in regulating the homeostasis of the internal environment and are closely associated with tumour progression as major mediators of cellular signalling. As a diverse and multifunctional group of proteins, the G protein signalling regulator (RGS) family was proven to be involved in the cellular transduction of GPCRs. Growing evidence has revealed dysregulation of RGS proteins as a common phenomenon and highlighted the key roles of these proteins in human cancers. Furthermore, their differential expression may be a potential biomarker for tumour diagnosis, treatment and prognosis. Most importantly, there are few systematic reviews on the functional/mechanistic characteristics and clinical application of RGS family members at present. In this review, we focus on the G-protein signalling regulator (RGS) family, which includes more than 20 family members. We analysed the classification, basic structure, and major functions of the RGS family members. Moreover, we summarize the expression changes of each RGS family member in various human cancers and their important roles in regulating cancer cell proliferation, stem cell maintenance, tumorigenesis and cancer metastasis. On this basis, we outline the molecular signalling pathways in which some RGS family members are involved in tumour progression. Finally, their potential application in the precise diagnosis, prognosis and treatment of different types of cancers and the main possible problems for clinical application at present are discussed. Our review provides a comprehensive understanding of the role and potential mechanisms of RGS in regulating tumour progression. Video Abstract.


Asunto(s)
Neoplasias , Proteínas RGS , Humanos , Transducción de Señal , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/metabolismo , Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
5.
J Immunol ; 211(11): 1656-1668, 2023 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-37850953

RESUMEN

Regulatory T cells (Tregs) are critical mediators of immune tolerance and play a diametric role in cancer and autoimmunity. Tumor-infiltrating Tregs are often associated with poor prognosis in solid tumors because their enrichment in the tumor microenvironment contributes to immunosuppression. Conversely, dysregulation in the Treg compartment can disrupt self-tolerance, leading to autoimmunity. In the present study, we describe what is, to our knowledge, a novel regulator of Tregs, the GTPase activator regulator of G protein 1 (RGS1), demonstrating that RGS1-deficient human Tregs show downregulation of Treg-associated genes and are less immunosuppressive. These RGS1-deficient Tregs exhibit perturbations to the FOXP3-c-MYC transcriptional axis and downstream metabolic and autophagy programs by shifting their energy demands toward glycolysis and rendering them less autophagic. Taken together, RGS1 may serve as an apical node of Treg function by regulating the FOXP3-c-MYC transcriptional axis, thereby providing a therapeutic rationale for targeting RGS1 for treatment of cancer and autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes , Neoplasias , Proteínas RGS , Humanos , Linfocitos T Reguladores , Enfermedades Autoinmunes/patología , Autoinmunidad , Neoplasias/patología , Autofagia/genética , Factores de Transcripción Forkhead/metabolismo , Microambiente Tumoral , Proteínas RGS/genética , Proteínas RGS/metabolismo
6.
Oncoimmunology ; 12(1): 2260620, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37781234

RESUMEN

Colorectal cancer (CRC) remains a leading cause of cancer-related mortality despite efforts to improve standard interventions. As CRC patients can benefit from immunotherapeutic strategies that incite effector T cell action, cancer vaccines represent a safe and promising therapeutic approach to elicit protective and durable immune responses against components of the tumor microenvironment (TME). In this study, we investigate the pre-clinical potential of a Listeria monocytogenes (Lm)-based vaccine targeting the CRC-associated vasculature. CRC survival and progression are reliant on functioning blood vessels to effectively mediate various metabolic processes and oxygenate underlying tissues. We, therefore, advance the strategy of initiating immunity in syngeneic mouse models against the endogenous pericyte antigen RGS5, which is a critical mediator of pathological vascularization. Overall, Lm-based vaccination safely induced potent anti-tumor effects that consisted of recruiting functional Type-1-associated T cells into the TME and reducing tumor blood vessel content. This study underscores the promising clinical potential of targeting RGS5 against vascularized tumors like CRC.


Asunto(s)
Neoplasias del Colon , Listeria monocytogenes , Listeria , Proteínas RGS , Ratones , Animales , Humanos , Pericitos , Neoplasias del Colon/prevención & control , Listeria monocytogenes/metabolismo , Vacunación , Microambiente Tumoral , Proteínas RGS/genética , Proteínas RGS/metabolismo
7.
Clin Exp Hypertens ; 45(1): 2271186, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-37879890

RESUMEN

Objective: Excessive proliferation and migration of pulmonary arterial smooth muscle cell (PASMC) is a core event of pulmonary hypertension (PH). Regulators of G protein signaling 10 (RGS10) can regulate cellular proliferation and cardiopulmonary diseases. We demonstrate whether RGS10 also serves as a regulator of PH.Methods: PASMC was challenged by hypoxia to induce proliferation and migration. Adenovirus carrying Rgs10 gene (Ad-Rgs10) was used for external expression of Rgs10. Hypoxia/SU5416 or MCT was used to induce PH. Right ventricular systolic pressure (RVSP) and right ventricular hypertrophy index (RVHI) were used to validate the establishment of PH model.Results: RGS10 was downregulated in hypoxia-challenged PASMC. Ad-Rgs10 significantly suppressed proliferation and migration of PASMC after hypoxia stimulus, while silencing RGS10 showed contrary effect. Mechanistically, we observed that phosphorylation of S6 and 4E-Binding Protein 1 (4EBP1), the main downstream effectors of mammalian target of rapamycin complex 1 (mTORC1) as well as phosphorylation of AKT, the canonical upstream of mTORC1 in hypoxia-induced PASMC were negatively modulated by RGS10. Both recovering mTORC1 activity and restoring AKT activity abolished these effects of RGS10 on PASMC. More importantly, AKT activation also abolished the inhibitory role of RGS10 in mTORC1 activity in hypoxia-challenged PASMC. Finally, we also observed that overexpression of RGS10 in vivo ameliorated pulmonary vascular wall thickening and reducing RVSP and RVHI in mouse PH model.Conclusion: Our findings reveal the modulatory role of RGS10 in PASMC and PH via AKT/mTORC1 axis. Therefore, targeting RGS10 may serve as a novel potent method for the prevention against PH."


Asunto(s)
Hipertensión Pulmonar , Proteínas RGS , Animales , Ratones , Proliferación Celular , Células Cultivadas , Proteínas de Unión al GTP/metabolismo , Proteínas de Unión al GTP/farmacología , Hipertensión Pulmonar/metabolismo , Hipertrofia Ventricular Derecha , Hipoxia/metabolismo , Mamíferos/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina/farmacología , Miocitos del Músculo Liso/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Arteria Pulmonar , Proteínas RGS/genética , Proteínas RGS/metabolismo , Proteínas RGS/farmacología
8.
FASEB J ; 37(8): e23064, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37440271

RESUMEN

Off target damage to vital organ systems is an unfortunate side effect of cancer chemotherapy and remains a major limitation to the use of these essential drugs in the clinic. Despite decades of research, the mechanisms conferring susceptibility to chemotherapy driven cardiotoxicity and hepatotoxicity remain unclear. In the livers of patients with a history of chemotherapy, we observed a twofold increase in expression of G protein regulator RGS7 and a corresponding decrease in fellow R7 family member RGS11. Knockdown of RGS7 via introduction of RGS7 shRNA via tail vein injection decreased doxorubicin-induced hepatic collagen and lipid deposition, glycogen accumulation, and elevations in ALT, AST, and triglycerides by approximately 50%. Surprisingly, a similar result could be achieved via introduction of RGS7 shRNA directly to the myocardium without impacting RGS7 levels in the liver directly. Indeed, doxorubicin-treated cardiomyocytes secrete the endocrine factors transforming growth factor ß1 (TGFß1) and TGFß superfamily binding protein follistatin-related protein 1 (FSTL1). Importantly, RGS7 overexpression in the heart was sufficient to recapitulate the impacts of doxorubicin on the liver and inhibition of TGFß1 signaling with the receptor blocker GW788388 ameliorated the effect of cardiac RGS7 overexpression on hepatic fibrosis, steatosis, oxidative stress, and cell death as well as the resultant elevation in liver enzymes. Together these data demonstrate that RGS7 controls both the release of TGFß1 from the heart and the profibrotic and pro-oxidant actions of TGFß1 in the liver and emphasize the functional significance of endocrine cardiokine signaling in the pathogenesis of chemotherapy drive multiorgan damage.


Asunto(s)
Proteínas Relacionadas con la Folistatina , Proteínas RGS , Humanos , Proteínas RGS/genética , Proteínas RGS/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Transducción de Señal/fisiología , Proteínas Portadoras/metabolismo , Hígado/metabolismo , Doxorrubicina/efectos adversos , Proteínas Relacionadas con la Folistatina/metabolismo
9.
Cell Signal ; 107: 110682, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37075876

RESUMEN

Regulator of protein signaling (RGS20) is a member of the RGS protein superfamily, which serve as key negative regulators of G protein-mediated signal transduction. Through their GTPase accelerating protein (GAP) activity, RGS proteins deactivate α-subunits of heterotrimeric G proteins. In addition, the majority of RGS proteins also have the ability to act through other, non-GAP related, functions. RGS20 is one of three members of the RZ subfamily, which all show selective GAP activity towards Gαz, however emerging data suggest that RGS20 can also regulate Gi/o-mediated signaling. While increased RGS20 expression is associated with the progression of multiple cancers, a large gap still exists relating to the mechanisms of RGS20 regulation and function. RGS20 contains a poly-cysteine string motif and a conserved cysteine in RGS domain, which are assumed to be palmitoylated. Palmitoylation, an important post-translational modification, plays an important role in cells by changing cellular functions of proteins. Consequently, the aim of this study was to confirm that RGS20 is palmitoylated and determine how palmitoylation affects its inhibition of Gαo-mediated signaling. We found a significant positive correlation between RGS20 palmitoylation and its association with active Gαo. We also showed that a conserved cysteine residue in the RGS domain is a critical site for its palmitoylation, with large impact on its association with Gαo. Palmitoylation on this site did not affect its GAP activity, however, it increased the inhibition of Gαo-mediated cAMP signaling. Altogether these data suggest that palmitoylation is a regulatory mechanism controlling RGS20 function, and that RGS20 can inhibit Gαo signaling through both GAP activity and non-GAP mechanisms.


Asunto(s)
Proteínas de Unión al GTP Heterotriméricas , Proteínas RGS , Proteínas RGS/metabolismo , Lipoilación , Cisteína/metabolismo , Transducción de Señal , Proteínas de Unión al GTP Heterotriméricas/metabolismo
10.
Curr Mol Med ; 23(9): 981-990, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37073154

RESUMEN

BACKGROUND: Osteoarthritis (OA) is a degenerative joint disease involving both cartilage and synovium. Activating transcription factor 3 (ATF3) and regulator of G protein signaling 1 (RGS1) have been reported to be up-regulated in OA. However, little is known regarding the relationship between these two genes and the mechanism of this relationship in OA development. Therefore, the present study explores the mechanism of ATF3-mediated RGS1 in the proliferation, migration, and apoptosis of synovial fibroblasts. METHODS: After the OA cell model was constructed with TGF-ß1 induction, human fibroblast-like synoviocytes (HFLSs) were transfected with ATF3 shRNA or RGS1 shRNA alone or co-transfected with ATF3 shRNA and pcDNA3.1-RGS1. Then, proliferation, migration, apoptosis, and the expression of ATF3, RGS1, α-SMA, BCL-2, caspase3, and cleaved-caspase3 were measured. Meanwhile, the potential relationship between ATF3 and RGS1 was predicted and validated. RESULTS AND DISCUSSION: Analysis of the GSE185059 dataset suggested that RGS1 was up-regulated in OA synovial fluid exosomes. Moreover, ATF3 and RGS1 were both highly expressed in TGF-ß1-induced HFLSs. Transfection of ATF3 shRNA or RGS1 shRNA significantly reduced proliferation and migration and promoted apoptosis of TGF- ß1-induced HFLSs. Mechanistically, ATF3 bound to the RGS1 promoter and elevated RGS1 expression. Silencing ATF3 repressed proliferation and migration and enhanced apoptosis of TGF-ß1-induced HFLSs by down-regulating RGS1. CONCLUSION: ATF3 binds to the RGS1 promoter and enhances RGS1 expression to accelerate cell proliferation and block cell apoptosis in TGF-ß1-induced synovial fibroblasts.


Asunto(s)
Proteínas RGS , Factor de Crecimiento Transformador beta1 , Humanos , Factor de Crecimiento Transformador beta1/farmacología , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Artroscopía , Fibroblastos/metabolismo , Apoptosis/genética , Proliferación Celular/genética , ARN Interferente Pequeño/metabolismo , Células Cultivadas , Proteínas RGS/genética , Proteínas RGS/metabolismo
11.
Ann Clin Lab Sci ; 53(2): 238-247, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37094862

RESUMEN

OBJECTIVE: Abdominal aortic aneurysm (AAA) is characterized by vascular smooth muscle cell (VSMC) injury. Circ_0000285 has been declared to drive cancer development, but its role in AAA remains unclear. We thus intended to disclose circ_0000285's role and molecular mechanism in AAA. METHODS: VSMCs were exposed to hydrogen peroxide (H2O2) to induce cell injury. Circ_0000285, miR-599, and regulator of G protein signaling 17 (RGS17) mRNA expressions were ascertained by conducting RT-qPCR assay while the levels of RGS17 protein was ascertained via western blotting. MiR-599's predicted binding with circ_0000285 and RGS17 were validated by means of the dual-luciferase reporter experiment. Cell proliferation was evaluated through the CCK-8 and EdU assays. Cell apoptosis was assessed via the caspase-3 activity assay. RESULTS: The AAA samples and H2O2-treated VSMCs manifested high expressions of circ_0000285 and RGS17 as well as a poor miR-599 expression. H2O2 treatment impaired the proliferation of VSMCs while stimulating their apoptosis. Circ_0000285 overexpression further repressed cell proliferation and enhanced apoptosis in H2O2-treated VSMCs while miR-599 enrichment partly reversed these effects. Circ_0000285 directly bound to miR-599, and miR-599 interacted with RGS17 3'UTR. RGS17 overexpression also suppressed cell proliferation and stimulated apoptosis in H2O2-treated VSMCs. Nevertheless, these effects were offset by miR-599 enrichment. CONCLUSION: Circ_0000285 governed the miR-599/RGS17 network to regulate H2O2-induced VSMC injuries, thereby promoting the development of AAA.


Asunto(s)
Aneurisma de la Aorta Abdominal , MicroARNs , Proteínas RGS , Humanos , MicroARNs/genética , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Peróxido de Hidrógeno/farmacología , Transducción de Señal , Aneurisma de la Aorta Abdominal/genética , Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/patología , Proliferación Celular/genética , Apoptosis , Proteínas RGS/metabolismo , Proteínas RGS/farmacología
12.
Cancer Biomark ; 36(4): 299-311, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36938729

RESUMEN

BACKGROUND: Regulatory T cells (Tregs) are central to determine immune response, thus targeting Tregs for immunotherapy is a promising strategy against tumor development and metastasis. OBJECTIVES: The objective of this study was to identify genes for targeting Tregs to improve the outcome of HCC. METHODS: We integrated expression data from different samples to remove batch effects and further applied embedding function in Scanpy to conduct sub-clustering of CD4+ T cells in HCC for each of two independent scRNA-seq data. The activity of transcription factors (TFs) was inferred by DoRothEA. Gene expression network analysis was performed in WGCNA R package. We finally used R packages (survminer and survival) to conduct survival analysis. Multiplex immunofluorescence analysis was performed to validate the result from bioinformatic analyses. RESULTS: We found that regulator of G protein signaling 1 (RGS1) expression was significantly elevated in Tregs compared to other CD4+ T cells in two independent public scRNA-seq datasets, and increased RGS1 predicted inferior clinical outcome of HCC patients. Multiplex immunofluorescence analysis supported that the higher expression of RGS1 in HCC Tregs in tumor tissue compared to it in adjacent tissue. Moreover, RGS1 expression in Tregs was positively correlated with the expression of marker genes of Tregs, C-X-C chemokine receptor 4 (CXCR4), and three CXCR4-dependent genes in both scRNA-seq and bulk RNA-seq data. We further identified that these three genes were selectively expressed in Tregs as compared to other CD4+ T cells. The activities of two transcription factors, recombination signal binding protein for immunoglobulin kappa J region (RBPJ) and yin yang 1 (YY1), were significantly different in HCC Tregs with RGS1 high and RGS1 low. CONCLUSIONS: Our findings suggested that RGS1 may regulate Treg function possibly through CXCR4 signaling and RGS1 could be a potential target to improve responses for immunotherapy in HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Proteínas RGS , Humanos , Carcinoma Hepatocelular/metabolismo , Proteínas de Unión al GTP , Neoplasias Hepáticas/metabolismo , Análisis de Expresión Génica de una Sola Célula , Linfocitos T Reguladores , Proteínas RGS/metabolismo
13.
Science ; 379(6639): 1352-1358, 2023 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-36996198

RESUMEN

Glycine is a major neurotransmitter involved in several fundamental neuronal processes. The identity of the metabotropic receptor mediating slow neuromodulatory effects of glycine is unknown. We identified an orphan G protein-coupled receptor, GPR158, as a metabotropic glycine receptor (mGlyR). Glycine and a related modulator, taurine, directly bind to a Cache domain of GPR158, and this event inhibits the activity of the intracellular signaling complex regulator of G protein signaling 7-G protein ß5 (RGS7-Gß5), which is associated with the receptor. Glycine signals through mGlyR to inhibit production of the second messenger adenosine 3',5'-monophosphate. We further show that glycine, but not taurine, acts through mGlyR to regulate neuronal excitability in cortical neurons. These results identify a major neuromodulatory system involved in mediating metabotropic effects of glycine, with implications for understanding cognition and affective states.


Asunto(s)
Glicina , Receptores Acoplados a Proteínas G , Receptores de Glicina , Glicina/metabolismo , Proteínas de Unión al GTP/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Glicina/química , Receptores de Glicina/genética , Receptores de Glicina/metabolismo , Transducción de Señal , Humanos , Células HEK293 , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Proteínas RGS/metabolismo , Dominios Proteicos
14.
Stem Cells ; 41(5): 493-504, 2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-36888549

RESUMEN

Regulator of G protein signaling 5 (RGS5) is a GTPase activator for heterotrimeric G-protein α-subunits, shown to be a marker of pericytes. Bone marrow stromal cell population (BMSCs) is heterogeneous. Populations of mesenchymal progenitors, cells supportive of hematopoiesis, and stromal cells regulating bone remodeling have been recently identified. Periosteal and bone marrow mesenchymal stem cells (MSCs) are participating in fracture healing, but it is difficult to distinguish the source of cells within the callus. Considering that perivascular cells exert osteoprogenitor potential, we generated an RGS5 transgenic mouse model (Rgs5-CreER) which when crossed with Ai9 reporter animals (Rgs5/Tomato), is suitable for lineage tracing during growth and post-injury. Flow cytometry analysis and histology confirmed the presence of Rgs5/Tomato+ cells within CD31+ endothelial, CD45+ hematopoietic, and CD31-CD45- mesenchymal/perivascular cells. A tamoxifen chase showed expansion of Rgs5/Tomato+ cells expressing osterix within the trabeculae positioned between mineralized matrix and vasculature. Long-term chase showed proportion of Rgs5/Tomato+ cells contributes to mature osteoblasts expressing osteocalcin. Following femoral fracture, Rgs5/Tomato+ cells are observed around newly formed bone within the BM cavity and expressed osterix and osteocalcin, while contribution within periosteum was low and limited to fibroblastic callus with very few positive chondrocytes. In addition, BM injury model confirmed that RGS5-Cre labels population of BMSCs expands during injury and participates in osteogenesis. Under homeostatic conditions, lineage-traced RGS5 cells within the trabecular area demonstrate osteoprogenitor capacity that in an injury model contributes to new bone formation primarily within the BM niche.


Asunto(s)
Callo Óseo , Proteínas RGS , Ratones , Animales , Osteocalcina/metabolismo , Callo Óseo/metabolismo , Callo Óseo/patología , Osteogénesis , Curación de Fractura/fisiología , Condrocitos/metabolismo , Ratones Transgénicos , Osteoblastos/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo
15.
Int J Oral Sci ; 15(1): 11, 2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36797232

RESUMEN

Tumor-associated macrophages (TAMs) play crucial roles in tumor progression and immune responses. However, mechanisms of driving TAMs to antitumor function remain unknown. Here, transcriptome profiling analysis of human oral cancer tissues indicated that regulator of G protein signaling 12 (RGS12) regulates pathologic processes and immune-related pathways. Mice with RGS12 knockout in macrophages displayed decreased M1 TAMs in oral cancer tissues, and extensive proliferation and invasion of oral cancer cells. RGS12 increased the M1 macrophages with features of increased ciliated cell number and cilia length. Mechanistically, RGS12 associates with and activates MYC binding protein 2 (MYCBP2) to degrade the cilia protein kinesin family member 2A (KIF2A) in TAMs. Our results demonstrate that RGS12 is an essential oral cancer biomarker and regulator for immunosuppressive TAMs activation.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Proteínas RGS , Ratones , Humanos , Animales , Macrófagos Asociados a Tumores/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello , Proteínas de Unión al GTP/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo , Cinesinas/metabolismo , Proteínas Represoras/metabolismo
16.
Exp Mol Med ; 55(1): 240-252, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36653442

RESUMEN

Production of estradiol (E2) by the placenta during human pregnancy ensures successful maintenance of placental development and fetal growth by stimulating trophoblast proliferation and the differentiation of cytotrophoblasts into syncytiotrophoblasts. Decreased levels of E2 are closely associated with obstetrical diseases such as preeclampsia (PE) in the clinic. However, the mechanisms underlying the inhibition of placental E2 biosynthesis remain poorly understood. Here, we report that regulator of G-protein signaling 2 (RGS2) affects E2 levels by regulating aromatase, a rate-limiting enzyme for E2 biosynthesis, by using human trophoblast-derived JEG-3 cells and human placental villus tissues. RGS2 enhanced the protein degradation of the transcription factor heart and neural crest derivatives expressed 1 (HAND1) by suppressing ubiquitin-specific protease 14 (USP14)-mediated deubiquitination of HAND1, resulting in the restoration of HAND1-induced trans-inactivation of the aromatase gene and subsequent increases in E2 levels. However, aromatase bound to RGS2 and repressed RGS2 GTPase activating protein (GAP) activity. Moreover, we observed a positive correlation between RGS2 and aromatase expression in clinical normal and preeclamptic placental tissues. Our results uncover a hitherto uncharacterized role of the RGS2-aromatase axis in the regulation of E2 production by human placental trophoblasts, which may pinpoint the molecular pathogenesis and highlight potential biomarkers for related obstetrical diseases.


Asunto(s)
Preeclampsia , Proteínas RGS , Humanos , Embarazo , Femenino , Trofoblastos/metabolismo , Placenta , Estradiol , Aromatasa/genética , Aromatasa/metabolismo , Línea Celular Tumoral , Preeclampsia/genética , Proteínas RGS/genética , Proteínas RGS/metabolismo , Ubiquitina Tiolesterasa/metabolismo
17.
Proc Natl Acad Sci U S A ; 120(1): e2213537120, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36574707

RESUMEN

Dose-limiting cardiotoxicity remains a major limitation in the clinical use of cancer chemotherapeutics. Here, we describe a role for Regulator of G protein Signaling 7 (RGS7) in chemotherapy-dependent heart damage, the demonstration for a functional role of RGS7 outside of the nervous system and retina. Though expressed at low levels basally, we observed robust up-regulation of RGS7 in the human and murine myocardium following chemotherapy exposure. In ventricular cardiomyocytes (VCM), RGS7 forms a complex with Ca2+/calmodulin-dependent protein kinase (CaMKII) supported by key residues (K412 and P391) in the RGS domain of RGS7. In VCM treated with chemotherapeutic drugs, RGS7 facilitates CaMKII oxidation and phosphorylation and CaMKII-dependent oxidative stress, mitochondrial dysfunction, and apoptosis. Cardiac-specific RGS7 knockdown protected the heart against chemotherapy-dependent oxidative stress, fibrosis, and myocyte loss and improved left ventricular function in mice treated with doxorubicin. Conversely, RGS7 overexpression induced fibrosis, reactive oxygen species generation, and cell death in the murine myocardium that were mitigated following CaMKII inhibition. RGS7 also drives production and release of the cardiokine neuregulin-1, which facilitates paracrine communication between VCM and neighboring vascular endothelial cells (EC), a maladaptive mechanism contributing to VCM dysfunction in the failing heart. Importantly, while RGS7 was both necessary and sufficient to facilitate chemotherapy-dependent cytotoxicity in VCM, RGS7 is dispensable for the cancer-killing actions of these same drugs. These selective myocyte-intrinsic and myocyte-extrinsic actions of RGS7 in heart identify RGS7 as an attractive therapeutic target in the mitigation of chemotherapy-driven cardiotoxicity.


Asunto(s)
Antineoplásicos , Cardiotoxicidad , Proteínas RGS , Animales , Humanos , Ratones , Antineoplásicos/efectos adversos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Cardiotoxicidad/metabolismo , Células Endoteliales/metabolismo , Fibrosis , Miocitos Cardíacos/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo
18.
Mol Pharmacol ; 103(1): 21-37, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36384958

RESUMEN

Regulators of G protein signaling (RGS) proteins modulate G protein-coupled receptor (GPCR) signaling by acting as negative regulators of G proteins. Genetic variants in RGS proteins are associated with many diseases, including cancers, although the impact of these mutations on protein function is uncertain. Here we analyze the RGS domains of 15 RGS protein family members using a novel bioinformatic tool that measures the missense tolerance ratio (MTR) using a three-dimensional (3D) structure (3DMTR). Subsequent permutation analysis can define the protein regions that are most significantly intolerant (P < 0.05) in each dataset. We further focused on RGS14, RGS10, and RGS4. RGS14 exhibited seven significantly tolerant and seven significantly intolerant residues, RGS10 had six intolerant residues, and RGS4 had eight tolerant and six intolerant residues. Intolerant and tolerant-control residues that overlap with pathogenic cancer mutations reported in the COSMIC cancer database were selected to define the functional phenotype. Using complimentary cellular and biochemical approaches, proteins were tested for effects on GPCR-Gα activation, Gα binding properties, and downstream cAMP levels. Identified intolerant residues with reported cancer-linked mutations RGS14-R173C/H and RGS4-K125Q/E126K, and tolerant RGS14-S127P and RGS10-S64T resulted in a loss-of-function phenotype in GPCR-G protein signaling activity. In downstream cAMP measurement, tolerant RGS14-D137Y and RGS10-S64T and intolerant RGS10-K89M resulted in change of function phenotypes. These findings show that 3DMTR identified intolerant residues that overlap with cancer-linked mutations cause phenotypic changes that negatively impact GPCR-G protein signaling and suggests that 3DMTR is a potentially useful bioinformatics tool for predicting functionally important protein residues. SIGNIFICANCE STATEMENT: Human genetic variant/mutation information has expanded rapidly in recent years, including cancer-linked mutations in regulator of G protein signaling (RGS) proteins. However, experimental testing of the impact of this vast catalogue of mutations on protein function is not feasible. We used the novel bioinformatics tool three-dimensional missense tolerance ratio (3DMTR) to define regions of genetic intolerance in RGS proteins and prioritize which cancer-linked mutants to test. We found that 3DMTR more accurately classifies loss-of-function mutations in RGS proteins than other databases thereby offering a valuable new research tool.


Asunto(s)
Neoplasias , Proteínas RGS , Humanos , Proteínas RGS/genética , Proteínas RGS/metabolismo , Transducción de Señal/genética , Proteínas de Unión al GTP/metabolismo , Mutación , Neoplasias/genética
19.
Antioxid Redox Signal ; 38(1-3): 137-159, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35521658

RESUMEN

Aims: The pathophysiological mechanism(s) underlying non-alcoholic fatty liver disease (NAFLD) have yet to be fully delineated and only a single drug, peroxisome proliferator-activated receptor (PPAR) α/γ agonist saroglitazar, has been approved. Here, we sought to investigate the role of Regulator of G Protein Signaling 7 (RGS7) in hyperlipidemia-dependent hepatic dysfunction. Results: RGS7 is elevated in the livers of NAFLD patients, particularly those with severe hepatic damage, pronounced insulin resistance, and high inflammation. In the liver, RGS7 forms a unique complex with transcription factor ATF3 and histone acetyltransferase Tip60, which is implicated in NAFLD. The removal of domains is necessary for ATF3/Tip60 binding compromises RGS7-dependent reactive oxygen species generation and cell death. Hepatic RGS7 knockdown (KD) prevented ATF3/Tip60 induction, and it provided protection against fibrotic remodeling and inflammation in high-fat diet-fed mice translating to improvements in liver function. Hyperlipidemia-dependent oxidative stress and metabolic dysfunction were largely reversed in RGS7 KD mice. Interestingly, saroglitazar failed to prevent RGS7/ATF3 upregulation but it did partially restore Tip60 levels. RGS7 drives the release of particularly tumor necrosis factor α (TNFα) from isolated hepatocytes, stellate cells and its depletion reverses steatosis, oxidative stress by direct TNFα exposure. Conversely, RGS7 overexpression in the liver is sufficient to trigger oxidative stress in hepatocytes that can be mitigated via TNFα inhibition. Innovation: We discovered a novel non-canonical function for an R7RGS protein, which usually functions to regulate G protein coupled receptor (GPCR) signaling. This is the first demonstration for a functional role of RGS7 outside the retina and central nervous system. Conclusion: RGS7 represents a potential novel target for the amelioration of NAFLD. Antioxid. Redox Signal. 38, 137-159.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Proteínas RGS , Animales , Ratones , Dieta Alta en Grasa , Inflamación/metabolismo , Hígado/metabolismo , Cirrosis Hepática/metabolismo , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Proteínas RGS/genética , Proteínas RGS/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
20.
Int J Mol Sci ; 23(21)2022 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-36362230

RESUMEN

G-protein-gated inwardly rectifying potassium (GIRK) channels are critical determinants of neuronal excitability. They have been proposed as potential targets to restore excitatory/inhibitory balance in acute amyloidosis models, where hyperexcitability is a hallmark. However, the role of GIRK signaling in transgenic mice models of Alzheimer's disease (AD) is largely unknown. Here, we study whether progressive amyloid-ß (Aß) accumulation in the hippocampus during aging alters GIRK channel expression in mutant ß-amyloid precursor protein (APPSw,Ind J9) transgenic AD mice. Additionally, we examine the impact of spatial memory training in a hippocampal-dependent task, on protein expression of GIRK subunits and Regulator of G-protein signaling 7 (RGS7) in the hippocampus of APPSw,Ind J9 mice. Firstly, we found a reduction in GIRK2 expression (the main neuronal GIRK channels subunit) in the hippocampus of 6-month-old APPSw,Ind J9 mice. Moreover, we found an aging effect on GIRK2 and GIRK3 subunits in both wild type (WT) and APPSw,Ind J9 mice. Finally, when 6-month-old animals were challenged to a spatial memory training, GIRK2 expression in the APPSw,Ind J9 mice were normalized to WT levels. Together, our results support the evidence that GIRK2 could account for the excitatory/inhibitory neurotransmission imbalance found in AD models, and training in a cognitive hippocampal dependent task may have therapeutic benefits of reversing this effect and lessen early AD deficits.


Asunto(s)
Enfermedad de Alzheimer , Proteínas RGS , Animales , Ratones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/terapia , Enfermedad de Alzheimer/metabolismo , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Hipocampo/metabolismo , Modelos Animales de Enfermedad , Ratones Transgénicos , Memoria Espacial , Proteínas RGS/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA