Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(9)2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38731904

RESUMEN

To maintain an optimal body content of phosphorus throughout postnatal life, variable phosphate absorption from food must be finely matched with urinary excretion. This amazing feat is accomplished through synchronised phosphate transport by myriads of ciliated cells lining the renal proximal tubules. These respond in real time to changes in phosphate and composition of the renal filtrate and to hormonal instructions. How they do this has stimulated decades of research. New analytical techniques, coupled with incredible advances in computer technology, have opened new avenues for investigation at a sub-cellular level. There has been a surge of research into different aspects of the process. These have verified long-held beliefs and are also dramatically extending our vision of the intense, integrated, intracellular activity which mediates phosphate absorption. Already, some have indicated new approaches for pharmacological intervention to regulate phosphate in common conditions, including chronic renal failure and osteoporosis, as well as rare inherited biochemical disorders. It is a rapidly evolving field. The aim here is to provide an overview of our current knowledge, to show where it is leading, and where there are uncertainties. Hopefully, this will raise questions and stimulate new ideas for further research.


Asunto(s)
Fosfatos , Humanos , Fosfatos/metabolismo , Animales , Reabsorción Renal , Riñón/metabolismo , Túbulos Renales Proximales/metabolismo
2.
Eur J Neurosci ; 57(2): 360-372, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36443250

RESUMEN

Regulator of G-protein signalling (RGS) proteins inhibit signalling by G-protein-coupled receptors (GPCRs). GPCRs mediate the functions of several important neurotransmitters and serve as targets of many anti-psychotics. RGS2, RGS4, RGS5 and RGS16 are located on chromosome 1q23.3-31, a locus found to be associated with schizophrenia. Although previous gene expression analysis detected down-regulation of RGS4 expression in brain samples of patients with schizophrenia, the results were not consistent. In the present study, we performed a systematic meta-analysis of differential RGS2, RGS4, RGS5 and RGS16 expression in Brodmann Area 10 (BA10) samples of patients with schizophrenia and from healthy controls. Two microarray datasets met the inclusion criteria (overall, 41 schizophrenia samples and 38 controls were analysed). RGS2 and RGS16 were found to be up-regulated in BA10 samples of individuals with schizophrenia, whereas no differential expression of RGS4 and RGS5 was detected. Analysis of dorso-lateral prefrontal cortex samples of the CommonMind Consortium (258 schizophrenia samples vs. 279 controls) further validated the results. Given their central role in inactivating G-protein-coupled signalling pathways, our results suggest that differential gene expression might lead to enhanced inactivation of G-protein signalling in schizophrenia. This, in turn, suggests that additional studies are needed to further explore the consequences of the differential expression we detected, this time at the protein and functional levels.


Asunto(s)
Regulación de la Expresión Génica , Corteza Prefrontal , Proteínas RGS , Esquizofrenia , Humanos , Expresión Génica , Perfilación de la Expresión Génica , Corteza Prefrontal/metabolismo , Proteínas RGS/genética , Esquizofrenia/genética , Regulación hacia Arriba
3.
Plant Cell Rep ; 42(12): 1987-2010, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37874341

RESUMEN

KEY MESSAGE: Nitrate-responsive transcriptomic, phenotypic and physiological analyses of rice RGA1 mutant revealed many novel RGA1-regulated genes/processes/traits related to nitrogen use efficiency, and provided robust genetic evidence of RGA1-regulation of NUE. Nitrogen (N) use efficiency (NUE) is important for sustainable agriculture. G-protein signalling was implicated in N-response/NUE in rice, but needed firm genetic characterization of the role of alpha subunit (RGA1). The knock-out mutant of RGA1 in japonica rice exhibited lesser nitrate-dose sensitivity than the wild type (WT), in yield and NUE. We, therefore, investigated its genomewide nitrate-response relative to WT. It revealed 3416 differentially expressed genes (DEGs), including 719 associated with development, grain yield and phenotypic traits for NUE. The upregulated DEGs were related to photosynthesis, chlorophyll, tetrapyrrole and porphyrin biosynthesis, while the downregulated DEGs belonged to cellular protein metabolism and transport, small GTPase signalling, cell redox homeostasis, etc. We validated 26 nitrate-responsive DEGs across functional categories by RT-qPCR. Physiological validation of nitrate-response in the mutant and the WT at 1.5 and 15 mM doses revealed higher chlorophyll and stomatal length but decreased stomatal density, conductance and transpiration. The consequent increase in photosynthesis and water use efficiency may have contributed to better yield and NUE in the mutant, whereas the WT was N-dose sensitive. The mutant was not as N-dose-responsive as the WT in shoot/root growth, productive tillers and heading date, but equally responsive as WT in total N and protein content. The RGA1 mutant was less impacted by higher N-dose or salt stress in terms of yield, protein content, photosynthetic performance, relative water content, water use efficiency and catalase activity. PPI network analyses revealed known NUE-related proteins as RGA1 interactors. Therefore, RGA1 negatively regulates N-dose sensitivity and NUE in rice.


Asunto(s)
Nitrógeno , Oryza , Nitrógeno/metabolismo , Nitratos/farmacología , Nitratos/metabolismo , Oryza/metabolismo , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Subunidades alfa de la Proteína de Unión al GTP/genética , Subunidades alfa de la Proteína de Unión al GTP/metabolismo , Perfilación de la Expresión Génica , Clorofila/metabolismo , Agua/metabolismo
4.
Oral Dis ; 29(4): 1802-1811, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-35122384

RESUMEN

OBJECTIVE: To explore the role of the Rgs10-associated nuclear factor (NF)-κB signalling pathway in periodontitis with rheumatoid arthritis. METHODS: Porphyromonas gingivalis and collagen were locally applied to mice to establish in vivo periodontitis and rheumatoid arthritis models, respectively. Both agents were administered together to establish the comorbid group. All models were treated with adeno-associated virus-green fluorescent protein (AAV-GFP) or adeno-associated virus small hairpin Rgs10 (AAV-sh-Rgs10). In vivo expression of Rgs10 and inflammatory cytokines was analysed, along with exploration of the NF-κB signalling pathway in lipopolysaccharide-stimulated mouse-derived RAW264.7 cells, with and without treatment of small interfering RNA (siRNA; Rgs10-Mus-MSS245072). RESULTS: In the comorbidity mouse group (mice with both periodontitis and rheumatoid arthritis), inhibition of Rgs10 exacerbated periodontitis, along with upregulation of phospho-RelA (pP65), tumour necrosis factor-α (TNF-α) and interleukin-6 (IL-6) expression in the NF-κB signalling pathway. Similarly, treatment of LPS-stimulated RAW264.7 cells with siRNA resulted in the inhibition of Rgs10, along with upregulation of pP65, TNF-α and IL-6 expression in vitro. CONCLUSION: Inhibition of Rgs10 in mice with periodontitis and rheumatoid arthritis can promote the progression of periodontitis, indicating the potential therapeutic role of Rgs10 in this condition.


Asunto(s)
Artritis Experimental , Artritis Reumatoide , Periodontitis , Proteínas RGS , Animales , Ratones , FN-kappa B/metabolismo , Interleucina-6 , Factor de Necrosis Tumoral alfa , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/metabolismo , Artritis Experimental/patología , ARN Interferente Pequeño/genética , Lipopolisacáridos/farmacología , Proteínas RGS/genética
5.
Int Endod J ; 56(7): 854-868, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37092953

RESUMEN

AIM: Apical periodontitis is a prevalent oral inflammatory disease that has recently been linked to transcription factor EB (TFEB)-mediated autophagy. Regulator of G-protein signalling 10 (RGS10) is reported to be an effective regulator of the immune system and inflammation. This study aimed to investigate the involvement of RGS10 during the development of apical periodontitis through the TFEB-mediated autophagy signalling pathway. METHODOLOGY: Sixty BALB/c mice were randomly divided into four groups of 15 mice for the in vivo experiment. Rgs10 was locally overexpressed through eight injections of an adeno-associated virus vector. The model of apical periodontitis was established 21 days following pulp exposure, and the mice were euthanized to obtain mandibles for analysis. Micro-computed tomography was employed to assess alveolar bone destruction, and the levels of Rgs10, TFEB-mediated autophagy signalling factors and inflammatory factors were measured using quantitative reverse transcription polymerase chain reactions, western blotting, enzyme-linked immunosorbent assays, immunofluorescence and immunohistochemistry. All experimental results were displayed as images or graphs. For the in vitro experiments, we employed small interfering RNA (siRNA) to silence Rgs10 expression in RAW 264.7 cells. The data were analysed via one-way anova or Mann-Whitney U test/Kruskal-Wallis test of variance, where p < .05 or U > 1.96 was considered statistically significant. RESULTS: Local overexpression of Rgs10 reduced alveolar bone destruction within the apical periodontitis lesion and significantly decreased macrophage infiltration (p < .05). Meanwhile, the expression of TFEB-mediated autophagy signalling factors was upregulated, along with a decrease in inflammatory factor expression (p < .05). Lipopolysaccharide-stimulated RAW 264.7 cells exhibited decreased Rgs10 expression and TFEB-mediated autophagy signalling. siRNA-mediated silencing of Rgs10 further suppressed autophagy and concomitantly upregulated inflammatory factors (p < .05). CONCLUSIONS: Collectively, the findings revealed that RGS10 suppresses the inflammatory response and bone destruction through TFEB-mediated autophagy in apical periodontitis.


Asunto(s)
Periodontitis Periapical , Proteínas RGS , Ratones , Animales , Microtomografía por Rayos X , Autofagia , Proteínas de Unión al GTP , ARN Interferente Pequeño
6.
Br Poult Sci ; 63(5): 613-620, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35522181

RESUMEN

1. The following study explored the expression and preliminary function of the RGS3 gene. The spatial and temporal expression patterns of the RGS3 gene were analysed in the ovarian stroma of Shendan No. 6 Green shell hens and Hy-line Brown hens at four time points (6, 28, 40, and 52 weeks old), as well as in various organs and follicles of Hy-line Brown hens.2. Based on the genomic and protein sequences of RGS3 in NCBI database, phylogenetic trees were constructed using MEGA-X. The protein interaction network was analysed using STRING. According to the results of protein-protein interaction network and pathways, the mRNA expression levels of RGS3 and three interaction proteins were explored by qRT-PCR in vitro.3. Spatio-temporal expression data revealed that RGS3 mRNA was expressed in all the organs tested, being highest in the hypothalamus. In different follicles, RGS3 mRNA was highly expressed in post-ovulatory follicles, followed by ovarian stroma and large white follicles. The expression levels of RGS3 mRNA in the ovarian stroma were significantly higher in Shendan No. 6 Green shell hens than that in the Hy-line Brown hens at all egg-laying stages.4. The phylogenetic tree results showed that ducks, geese and chickens had higher homology based on the genomic and protein sequence of RGS3. Moreover, chicken RGS3 interacted with GSK3B, RAF1 and BRAF based on STRING prediction. In vitro follicle stimulating hormone (FSH) treatment showed that mRNA expression levels of RGS3 and those of its predicted interacting proteins BRAF and GSK3B decreased with increasing FSH concentration. The results suggested that RGS3 responds to FSH and may play an important role in the regulation of follicular development in chicken.


Asunto(s)
Pollos , Folículo Ovárico , Animales , Femenino , Pollos/fisiología , Folículo Ovárico/metabolismo , Filogenia , Proteínas Proto-Oncogénicas B-raf/metabolismo , Hormona Folículo Estimulante/metabolismo , ARN Mensajero/metabolismo
7.
Mol Genet Genomics ; 295(2): 357-371, 2020 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-31776761

RESUMEN

Females and males differ substantially in various neuronal functions in divergent, sexually dimorphic animal species, including humans. Despite its developmental, physiological and medical significance, understanding the molecular mechanisms by which sex-specific differences in the anatomy and operation of the nervous system are established remains a fundamental problem in biology. Here, we show that in Caenorhabditis elegans (nematodes), the global sex-determining factor TRA-1 regulates food leaving (mate searching), male mating and adaptation to odorants in a sex-specific manner by repressing the expression of goa-1 gene, which encodes the Gα(i/o) subunit of heterotrimeric G (guanine-nucleotide binding) proteins triggering physiological responses elicited by diverse neurotransmitters and sensory stimuli. Mutations in tra-1 and goa-1 decouple behavioural patterns from the number of X chromosomes. TRA-1 binds to a conserved binding site located in the goa-1 coding region, and downregulates goa-1 expression in hermaphrodites, particularly during embryogenesis when neuronal development largely occurs. These data suggest that the sex-determination machinery is an important modulator of heterotrimeric G protein-mediated signalling and thereby various neuronal functions in this organism and perhaps in other animal phyla.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Proteínas de Unión al ADN/genética , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Neuronas/metabolismo , Factores de Transcripción/genética , Animales , Sitios de Unión/genética , Caenorhabditis elegans/crecimiento & desarrollo , Desarrollo Embrionario/genética , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Masculino , Mutación/genética , Procesos de Determinación del Sexo/genética , Cromosoma X/genética
8.
Pharmacol Res ; 160: 105148, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32858121

RESUMEN

Neuropathic pain, a specific type of chronic pain resulting from persistent nervous tissue lesions, is a debilitating condition that affects about 7% of the population. This condition remains particularly difficult to treat because of the poor understanding of its underlying mechanisms. Drugs currently used to alleviate this chronic pain syndrome are of limited benefit due to their lack of efficacy and the elevated risk of side effects, especially after a prolonged period of treatment. Although drugs targeting G protein-coupled receptors (GPCR) also have several limitations, such as progressive loss of efficacy due to receptor desensitization or unavoidable side effects due to wide receptor distribution, the identification of several molecular partners that contribute to the fine-tuning of receptor activity has raised new opportunities for the development of alternative therapeutic approaches. Regulators of G protein signalling (RGS) act intracellularly by influencing the coupling process and activity of G proteins, and are amongst the best-characterized physiological modulators of GPCR. Changes in RGS expression have been documented in a range of models of neuropathic pain, or after prolonged treatment with diverse analgesics, and could participate in altered pain processing as well as impaired physiological or pharmacological control of nociceptive signals. The present review summarizes the experimental data that implicates RGS in the development of pain with focus on the pathological mechanisms of neuropathic pain, including the impact of neuropathic lesions on RGS expression and, reciprocally, the influence of modifying RGS on GPCRs involved in the modulation of nociception as well as on the outcome of pain. In this context, we address the question of the relevance of RGS as promising targets in the treatment of neuropathic pain.


Asunto(s)
Proteínas de Unión al GTP/efectos de los fármacos , Neuralgia/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Animales , Dolor Crónico , Proteínas de Unión al GTP/agonistas , Humanos , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/efectos de los fármacos
9.
Int J Mol Sci ; 22(1)2020 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-33374915

RESUMEN

We and others have previously identified signalling pathways associated with the adenosine A1 receptor (A1R) as important regulators of cellular responses to injury in the cochlea. We have shown that the "post-exposure" treatment with adenosine A1R agonists confers partial protection against acoustic trauma and other forms of sensorineural hearing loss (SNHL). The aim of this study was to determine if increasing A1R responsiveness to endogenous adenosine would have the same otoprotective effect. This was achieved by pharmacological targeting of the Regulator of G protein Signalling 4 (RGS4). RGS proteins inhibit signal transduction pathways initiated by G protein-coupled receptors (GPCR) by enhancing GPCR deactivation and receptor desensitisation. A molecular complex between RGS4 and neurabin, an intracellular scaffolding protein expressed in neural and cochlear tissues, is the key negative regulator of A1R activity in the brain. In this study, Wistar rats (6-8 weeks) were exposed to traumatic noise (110 dBSPL, 8-16 kHz) for 2 h and a small molecule RGS4 inhibitor CCG-4986 was delivered intratympanically in a Poloxamer-407 gel formulation for sustained drug release 24 or 48 h after noise exposure. Intratympanic administration of CCG-4986 48 h after noise exposure attenuated noise-induced permanent auditory threshold shifts by up to 19 dB, whilst the earlier drug administration (24 h) led to even better preservation of auditory thresholds (up to 32 dB). Significant improvement of auditory thresholds and suprathreshold responses was linked to improved survival of sensorineural tissues and afferent synapses in the cochlea. Our studies thus demonstrate that intratympanic administration of CCG-4986 can rescue cochlear injury and hearing loss induced by acoustic overexposure. This research represents a novel paradigm for the treatment of various forms of SNHL based on regulation of GPCR.


Asunto(s)
Pérdida Auditiva Provocada por Ruido/prevención & control , Pérdida Auditiva Sensorineural/prevención & control , Proteínas RGS/antagonistas & inhibidores , Sulfonamidas/farmacología , Animales , Umbral Auditivo/efectos de los fármacos , Cóclea/efectos de los fármacos , Cóclea/metabolismo , Potenciales Evocados Auditivos del Tronco Encefálico/efectos de los fármacos , Células Ciliadas Auditivas/efectos de los fármacos , Pérdida Auditiva Provocada por Ruido/metabolismo , Pérdida Auditiva Sensorineural/metabolismo , Masculino , Proteínas de Microfilamentos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas RGS/metabolismo , Ratas Wistar , Receptor de Adenosina A1/metabolismo , Transducción de Señal/efectos de los fármacos
10.
Biochem Biophys Res Commun ; 499(2): 143-149, 2018 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-29534968

RESUMEN

Regulator of G-protein signalling 5 (RGS5) is, highly expressed in different cell types of the adult human heart, and it is a negative regulator of G protein-mediated signalling that inactivates Gα(q) and Gα(i) and thereby inhibits many signalling pathways. However, the critical role of RGS5 in the pathology of myocardial infarction (MI) remains unexplored. Here, an in vitro MI model, induced by the permanent ligation of the left anterior descending coronary artery, was used with the isolated hearts of wild type (WT) and RGS5-knockout (KO) mice. Our results showed that the loss of RGS5 decreased the post-MI survival rate and left ventricular (LV) function and increased the infarct size. Additionally, the RGS5 knockout mice exhibited greater inflammation, apoptosis, and ventricular remodelling compared with WT-MI mice. Mechanistically, RGS5 loss activated the pathological response mainly by affecting the NF-κB and MAPK signalling pathways. Therefore, our data strongly indicate that RGS5 is a novel modulator of pathological progression after MI that functions NF-κB and MAPK signalling.


Asunto(s)
Sistema de Señalización de MAP Quinasas , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , FN-kappa B/metabolismo , Proteínas RGS/metabolismo , Remodelación Ventricular , Animales , Muerte Celular , Eliminación de Gen , Inflamación/complicaciones , Inflamación/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/enzimología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología
11.
Plant Biotechnol J ; 2018 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-29431919

RESUMEN

Verticillium wilt (VW), caused by soil-borne fungi of the genus Verticillium, is a serious disease affecting a wide range of plants and leading to a constant and major challenge to agriculture worldwide. Cotton (Gossypium hirsutum) is the world's most important natural textile fibre and oil crop. VW of cotton is a highly devastating vascular disease; however, few resistant germplasms have been reported in cotton. An increasing number of studies have shown that RNA interference (RNAi)-based host-induced gene silencing (HIGS) is an effective strategy for improving plant resistance to pathogens by silencing genes essential for the pathogenicity of these pathogens. Here, we have identified and characterized multifunctional regulators of G protein signalling (RGS) in the Verticillium dahliae virulence strain, Vd8. Of eight VdRGS genes, VdRGS1 showed the most significant increase in expression in V. dahliae after treating with the roots of cotton seedlings. Based on the phenotype detection of VdRGS1 deletion and complementation mutants, we found that VdRGS1 played crucial roles in spore production, hyphal development, microsclerotia formation and pathogenicity. Tobacco rattle virus-mediated HIGS in cotton plants silenced VdRGS1 transcripts in invaded V. dahliae strains and enhanced broad-spectrum resistance to cotton VW. Our data demonstrate that VdRGS1 is a conserved and essential gene for V. dahliae virulence. HIGS of VdRGS1 provides effective control against V. dahliae infection and could obtain the durable disease resistance in cotton and in other VW-susceptible host crops by developing the stable transformants.

12.
Cell Biochem Funct ; 35(6): 334-338, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28845525

RESUMEN

Recent evidence suggests that adventitial fibroblasts (AFs) are crucially implicated in atherosclerosis. However, the mechanisms by which AFs are dysfunctional and contribute to atherosclerosis remain unclear. This study aimed to investigate the role of regulator of G-protein signalling 3 (RGS3) in the regulation of AFs using apoE knockout mouse as the model. Pathological changes in aortic arteries of apoE knockout mice fed with hyperlipid diet were examined by Movat staining. The expression of RGS3, α-SMA, TGF-ß1, Smad2, and Smad3 in the adventitia was detected by immunohistochemistry. Adventitial fibroblasts were isolated from aortic arteries of apoE knockout mice and infected with RGS3 overexpression lentivirus or empty lentivirus. The expression of RGS3, α-SMA, TGF-ß1, Smad2, and Smad3 in AFs was detected by real-time polymerase chain reaction and Western blot analysis. We found that hyperlipidic diet caused significant aortic intima thickening and atherosclerotic plaques in 15-week-old apoE knockout mice. Compared to wild-type mice, RGS3 expression was lower while α-SMA, TGF-ß1, Smad2, and Smad3 expression was higher in the adventitia of apoE knockout mice. In addition, lentivirus mediated overexpression of RGS3 caused decreased expression of α-SMA, TGF-ß1, Smad2, and Smad3 in AFs derived from apoE(-/-) mice. In conclusion, these results suggest that RGS3 may provide protection against pathological changes of AFs and the development of atherosclerosis by inhibiting TGF-ß1/Smad signalling. RGS3 may be a potential therapeutic target for atherosclerosis.


Asunto(s)
Proteínas RGS/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Actinas/genética , Actinas/metabolismo , Animales , Aorta/citología , Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Células Cultivadas , Dieta Alta en Grasa , Regulación hacia Abajo , Fibroblastos/citología , Fibroblastos/metabolismo , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Inmunohistoquímica , Lentivirus/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas RGS/genética , ARN Mensajero/metabolismo , Transducción de Señal , Proteína Smad2/genética , Proteína smad3/genética , Factor de Crecimiento Transformador beta1/genética
13.
Adv Exp Med Biol ; 1002: 209-225, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28600788

RESUMEN

Mitosis is a process requiring strict spatial organization of cellular components. In particular, the orientation of the mitotic spindle with respect to the tissue defines the division plane. In turn, the orientation of cell division can regulate tissue morphology or the fate of daughter cells. While we have learned much about the mechanisms of mitotic spindle orientation, recent studies suggest that the proteins implicated can also play important roles in post-mitotic cells. Interestingly, post-mitotic protein function often involves polarizing the cell cytoskeleton during differentiation, mirroring its ability to orient the mitotic spindle during division. This review focuses on alternative functions of the spindle orientation machinery after division, when the cell undergoes a specialization process associated with differentiation or mature function, and discusses diseases associated to those alternative functions.


Asunto(s)
Diferenciación Celular , Mitosis , Huso Acromático/patología , Animales , Humanos , Fenotipo , Transducción de Señal , Huso Acromático/metabolismo
14.
Neurobiol Dis ; 94: 95-105, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27288154

RESUMEN

Parkinson's disease (PD) is a neurodegenerative disease where the degeneration of the nigrostriatal pathway leads to specific motor deficits. There is an unmet medical need for regenerative treatments that stop or reverse disease progression. Several growth factors have been investigated in clinical trials to restore the dopaminergic nigrostriatal pathway damaged in PD. Platelet-derived growth factor-BB (PDGF-BB), a molecule that recruits pericytes to stabilize microvessels, was recently investigated in a phase-1 clinical trial, showing a dose-dependent increase in dopamine transporter binding in the putamen of PD patients. Interestingly, evidence is accumulating that PD is paralleled by microvascular changes, however, whether PDGF-BB modifies pericytes in PD is not known. Using a pericyte reporter mouse strain, we investigate the functional and restorative effect of PDGF-BB in a partial 6-hydroxydopamine medial forebrain bundle lesion mouse model of PD, and whether this restorative effect is accompanied by changes in pericyte features. We demonstrate that a 2-week treatment with PDGF-BB leads to behavioural recovery using several behavioural tests, and partially restores the nigrostriatal pathway. Interestingly, we find that pericytes are activated in the striatum of PD lesioned mice and that these changes are reversed by PDGF-BB treatment. The modulation of brain pericytes may contribute to the PDGF-BB-induced neurorestorative effects, PDGF-BB allowing for vascular stabilization in PD. Pericytes might be a new cell target of interest for future regenerative therapies.


Asunto(s)
Actividad Motora/efectos de los fármacos , Enfermedad de Parkinson/metabolismo , Pericitos/efectos de los fármacos , Proteínas Proto-Oncogénicas c-sis/farmacología , Animales , Becaplermina , Cuerpo Estriado/efectos de los fármacos , Cuerpo Estriado/metabolismo , Modelos Animales de Enfermedad , Dopamina/metabolismo , Masculino , Haz Prosencefálico Medial/efectos de los fármacos , Haz Prosencefálico Medial/metabolismo , Ratones Transgénicos , Actividad Motora/fisiología , Oxidopamina/farmacología , Enfermedad de Parkinson/patología , Pericitos/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo
15.
Pharmacol Res ; 105: 54-61, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26773203

RESUMEN

Atrial fibrillation is the commonest cardiac arrhythmia and leads to significant clinical morbidity and mortality. It has a complex pathophysiology but is often initiated by atrial ectopic beats and because of atrial remodelling once it occurs it can become established. Thus therapeutic interventions designed to prevent the initial occurrence of the arrhythmia are particularly needed. At the cellular level, these ectopic beats arise because of abnormal calcium release events from the sarcoplasmic reticulum leading to an inward current mediated by the sodium-calcium exchanger. There has been considerable interest in this over the last few years largely focused on the ryanodine receptor and related signalling pathways. However, atrial myocytes also possess a well-developed inositol trisphosphate (IP3) dependent calcium release system and this has been less studied. In this review we focus on pathways and molecules that couple via the Gq\11 family of G-proteins including regulators of G-protein signalling that may influence IP3 mediated calcium release and atrial fibrillation.


Asunto(s)
Fibrilación Atrial/metabolismo , Calcio/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Fosfatos de Inositol/metabolismo , Transducción de Señal , Animales , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/patología , Descubrimiento de Drogas , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/metabolismo , Atrios Cardíacos/patología , Humanos , Terapia Molecular Dirigida , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Transducción de Señal/efectos de los fármacos
16.
Int J Mol Med ; 53(3)2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38214344

RESUMEN

Osteocyte function is critical for metabolism, remodelling and regeneration of bone tissue. In the present study, the roles of regulator of G protein signalling 18 (RGS18) were assessed in the regulation of osteocyte proliferation and bone formation. Target genes and signalling pathways were screened using the Gene Expression Omnibus (GEO) database and Gene Set Enrichment Analysis (GSEA). The function of RGS18 and the associated mechanisms were analysed by Cell Counting Kit 8 assay, 5­ethynyl­2'­deoxyuridine assay, flow cytometry, reverse transcription­quantitative PCR, western blotting and immunostaining. Overlap analysis of acutely injured subjects (AIS) and healthy volunteers (HVs) from the GSE93138 and GSE93215 datasets of the GEO database identified four genes: KIAA0825, ANXA3, RGS18 and LIPN. Notably, RGS18 was more highly expressed in peripheral blood samples from AIS than in those from HVs. Furthermore, RGS18 overexpression promoted MLO­Y4 and MC3T3­E1 cell viability, proliferation and S­phase arrest, but inhibited apoptosis by suppressing caspase­3/9 cleavage. Silencing RGS18 exerted the opposite effects. GSEA of GSE93138 revealed that RGS18 has the ability to regulate MAPK signalling. Treatment with the MEK1/2 inhibitor PD98059 reversed the RGS18 overexpression­induced osteocyte proliferation, and treatment with the ERK1/2 activator 12­O­tetradecanoylphorbol­13­acetate reversed the effects of RGS18 silencing on osteocyte proliferation. In conclusion, RGS18 may contribute to osteocyte proliferation and bone fracture healing via activation of ERK signalling.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular , Osteocitos , Proteínas RGS , Humanos , Apoptosis/genética , Proliferación Celular/genética , Proteínas de Unión al GTP , Transducción de Señal , Animales , Ratones , Células 3T3 , Proteínas RGS/genética
17.
Cells ; 13(13)2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38994966

RESUMEN

Fluorescence resonance energy transfer (FRET) biosensors have proven to be an indispensable tool in cell biology and, more specifically, in the study of G-protein signalling. The best method of measuring the activation status or FRET state of a biosensor is often fluorescence lifetime imaging microscopy (FLIM), as it does away with many disadvantages inherent to fluorescence intensity-based methods and is easily quantitated. Despite the significant potential, there is a lack of reliable FLIM-FRET biosensors, and the data processing and analysis workflows reported previously face reproducibility challenges. Here, we established a system in live primary mouse pancreatic ductal adenocarcinoma cells, where we can detect the activation of an mNeonGreen-Gαi3-mCherry-Gγ2 biosensor through the lysophosphatidic acid receptor (LPAR) with 2-photon time-correlated single-photon counting (TCSPC) FLIM. This combination gave a superior signal to the commonly used mTurquoise2-mVenus G-protein biosensor. This system has potential as a platform for drug screening, or to answer basic cell biology questions in the field of G-protein signalling.


Asunto(s)
Técnicas Biosensibles , Transferencia Resonante de Energía de Fluorescencia , Animales , Transferencia Resonante de Energía de Fluorescencia/métodos , Ratones , Técnicas Biosensibles/métodos , Proteínas de Unión al GTP/metabolismo , Humanos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Línea Celular Tumoral , Receptores del Ácido Lisofosfatídico/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología
18.
Clin Respir J ; 18(1): e13712, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38081176

RESUMEN

INTRODUCTION: Regulator of G protein signalling 1 (RGS1) closely regulates malignant phenotypes and tumour immunity in several cancers, while its clinical value in nonsmall cell lung cancer (NSCLC) is by far rarely reported. Consequently, this study aimed to explore the linkage of blood RGS1 with clinical features and prognosis in surgical NSCLC patients. METHODS: Two-hundred and ten surgical NSCLC patients were consecutively enrolled in this study, whose RGS1 in peripheral blood mononuclear cells was determined before treatment via reverse transcriptional-quantitative polymerase chain reaction. Additionally, the blood RGS1 was also collected from 30 healthy controls (HCs). RESULTS: Blood RGS1 was increased in NSCLC patients compared with HCs (P < 0.001). Elevated blood RGS1 was related to lymph node (LYN) metastasis (P = 0.001), higher tumour-nodes-metastasis (TNM) stage (P = 0.004), neoadjuvant chemotherapy administration (P = 0.044), shortened accumulative disease-free survival (DFS) (P = 0.008) and overall survival (OS) (P = 0.013) in NSCLC patients. A multivariate Cox's regression analysis showed that blood RGS1 high expression could independently reflect shortened DFS (hazard ratio = 1.499, P = 0.023), whereas it could not independently predict OS (P > 0.050). Furthermore, blood RGS1 high expression was associated with shortened OS (P = 0.020) in patients with neoadjuvant therapy and with worse DFS (P = 0.028) and OS (P = 0.026) in patients with adjuvant therapy, while blood RGS1 was not linked with DFS or OS in patients without neoadjuvant or adjuvant therapy (all P > 0.050). CONCLUSION: Elevated blood RGS1 correlates with LYN metastasis, neoadjuvant chemotherapy administration, worse DFS and OS, which might serve as a useful prognostic biomarker for surgical NSCLC patients.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/metabolismo , Pronóstico , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/patología , Supervivencia sin Enfermedad , Proteínas de Unión al GTP/metabolismo , Biomarcadores , Estadificación de Neoplasias
19.
J Exp Bot ; 64(14): 4517-27, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24163402

RESUMEN

A rice cDNA, OsDEP1, encoding a highly cysteine (Cys)-rich G protein γ subunit, was initially identified as it conferred cadmium (Cd) tolerance on yeast cells. Of the 426 aa constituting OsDEP1, 120 are Cys residues (28.2%), of which 88 are clustered in the C-terminal half region (aa 170-426). To evaluate the independent effects of these two regions, two truncated versions of the OsDEP1-expressing plasmids pOsDEP1(1-169) and pOsDEP1(170-426) were used to examine their effects on yeast Cd tolerance. Although OsDEP1(170-426) conferred a similar level of Cd tolerance as the intact OsDEP1, OsDEP1(1-169) provided no such tolerance, indicating that the tolerance effect is localized to the aa 170-426 C-terminal peptide region. The Cd responses of transgenic Arabidopsis plants constitutively expressing OsDEP1, OsDEP1(1-169) or OsDEP1(170-426), were similar to the observations in yeast cells, with OsDEP1 and OsDEP1(170-426) transgenic plants displaying Cd tolerance but OsDEP1(1-169) plants showing no such tolerance. In addition, a positive correlation between the transcript levels of OsDEP1 or OsDEP1(170-426) in the transgenics and the Cd content of these plants upon Cd application was observed. As several Arabidopsis loss-of-function heterotrimeric G protein ß and γ subunit gene mutants did not show differences in their Cd sensitivity compared with wild-type plants, we propose that the Cys-rich region of OsDEP1 may function directly as a trap for Cd ions.


Asunto(s)
Adaptación Fisiológica/efectos de los fármacos , Cadmio/toxicidad , Cisteína/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Oryza/metabolismo , Proteínas de Plantas/metabolismo , Saccharomyces cerevisiae/fisiología , Arabidopsis/efectos de los fármacos , Arabidopsis/genética , Cobre/toxicidad , Subunidades gamma de la Proteína de Unión al GTP/química , Mutación/genética , Oryza/efectos de los fármacos , Oryza/fisiología , Proteínas de Plantas/química , Plantas Modificadas Genéticamente , Estructura Terciaria de Proteína , Reproducibilidad de los Resultados , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/efectos de los fármacos
20.
Arab J Gastroenterol ; 21(4): 237-245, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32928707

RESUMEN

BACKGROUND AND STUDY AIMS: Regulator of G-protein signalling 3 (RGS3) plays a pivotal role in Wnt signalling and epithelial-mesenchymal transition. RGS3 overexpression in gastric cancer suggests that RGS3 and its regulators have the potential to serve as therapeutic targets for gastric cancer. Therefore, we aimed to investigate the roles of RGS3 and its regulator microRNA-133a in gastric cancer tumorigenesis. MATERIAL AND METHODS: mRNA and protein expression levels of RGS3 in 107 paired human gastric cancer tissues and gastric cancer cells were examined using qRT-PCR and immunoblotting, respectively. The relationship between RGS3/microRNA-133a expression and clinicopathological characteristics was assessed using t-test. TargetScan, miRanda and MicroCosm Targets were employed to predict the binding site on the 3'-untranslated region of RGS3 that is targeted by microRNA-133a. Moreover, dual-luciferase reporter assay was performed to validate target prediction. microRNA-133a expression level in gastric cancer tissues and cell lines was determined by qRT-PCR. Finally, the proliferation activity of gastric cancer cells was evaluated using Cell Counting Kit-8 and bromodeoxyuridine incorporation assays. RESULTS: RGS3 expression level markedly increased in both gastric cancer tissues and cells compared with that in the corresponding normal tissues and cells. However, microRNA-133a expression level markedly decreased in gastric cancer tissues and cells and was negatively correlated with RGS3 expression. Higher RGS3 and lower microRNA-133a expression levels were associated with a larger tumour size, lymph node metastasis, local invasion and advanced tumour-node-metastasis stage in gastric cancer. Dual-luciferase reporter assay verified that microRNA-133a targeted RGS3 via mRNA 3'-untranslated region binding. Finally, microRNA-133a inhibited gastric cancer cell proliferation, whereas RGS3 overexpression attenuated this inhibitory effect. CONCLUSION: MicroRNA-133a is a regulator of RGS3 in gastric cancer and the microRNA-133a-RGS3 axis possibly participates in the malignant progression of gastric cancer.


Asunto(s)
Proteínas RGS/genética , Neoplasias Gástricas , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs , Neoplasias Gástricas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA