Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Base de datos
Tipo del documento
Intervalo de año de publicación
1.
Discov Oncol ; 15(1): 135, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38679635

RESUMEN

Rectal adenocarcinoma (READ) is a common malignant tumor of the digestive tract. Growing studies have confirmed Ras GTPase-activating proteins are involved in the progression of several tumors. This study aimed to explore the expression and function of Ras GTPase-activating proteins in READ. In this study, we analyzed RNA sequencing data from 165 patients with READ and 789 normal tissue samples, identifying 5603 differentially expressed genes (DEGs), including 2937 upregulated genes and 2666 downregulated genes. Moreover, we also identified two dysregulated genes, RASA4 and SYNGAP1, among six Ras GTPase-activating proteins. High NF1 expression was associated with longer overall survival, while high SYNGAP1 expression showed a trend towards extended overall survival. Further analysis revealed the mutation frequency and copy number variations of Ras GTPase-activating proteins in various cancer samples. Additionally, DNA methylation analysis demonstrated a negative correlation between DNA methylation of Ras GTPase-activating proteins and their expression. Moreover, among Ras GTPase-activating proteins, we focused on SYNGAP1, and experimental validation confirmed that the overexpression of SYNGAP1 in READ significantly suppressed READ cell proliferation and increased apoptosis via regulating the Wnt/ß-Catenin signaling pathway. These findings underscored the potential significance of SYNGAP1 in READ and provide new insights for further research and treatment.

2.
Front Mol Neurosci ; 16: 1125277, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37063367

RESUMEN

Rho GTPases family are considered to be molecular switches that regulate various cellular processes, including cytoskeleton remodeling, cell polarity, synaptic development and maintenance. Accumulating evidence shows that Rho GTPases are involved in neuronal development and brain diseases, including substance dependence. However, the functions of Rho GTPases in substance dependence are divergent and cerebral nuclei-dependent. Thereby, comprehensive integration of their roles and correlated mechanisms are urgently needed. In this review, the molecular functions and regulatory mechanisms of Rho GTPases and their regulators such as GTPase-activating proteins (GAPs) and guanine nucleotide exchange factors (GEFs) in substance dependence have been reviewed, and this is of great significance for understanding their spatiotemporal roles in addictions induced by different addictive substances and in different stages of substance dependence.

3.
Eur J Med Chem ; 248: 115104, 2023 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-36641861

RESUMEN

To search more therapeutic strategies for Ras-mutant tumors, regulators of the Ras superfamily involved in the GTP/GDP (guanosine triphosphate/guanosine diphosphate) cycle have been well concerned for their anti-tumor potentials. GTPase activating proteins (GAPs) provide the catalytic group necessary for the hydrolysis of GTPs, which accelerate the switch by cycling between GTP-bound active and GDP-bound inactive forms. Inactivated GAPs lose their function in activating GTPase, leading to the continuous activation of downstream signaling pathways, uncontrolled cell proliferation, and eventually carcinogenesis. A growing number of evidence has shown the close link between GAPs and human tumors, and as a result, GAPs are believed as potential anti-tumor targets. The present review mainly summarizes the critically important role of GAPs in human tumors by introducing the classification, function and regulatory mechanism. Moreover, we comprehensively describe the relationship between dysregulated GAPs and the certain type of tumor. Finally, the current status, research progress, and clinical value of GAPs as therapeutic targets are also discussed, as well as the challenges and future direction in the cancer therapy.


Asunto(s)
Neoplasias , Proteínas Activadoras de ras GTPasa , Humanos , Proteínas Activadoras de ras GTPasa/metabolismo , Proteínas Activadoras de GTPasa , GTP Fosfohidrolasas , Guanosina Trifosfato/metabolismo , Guanosina Difosfato/metabolismo , Neoplasias/tratamiento farmacológico
4.
FEBS Lett ; 597(6): 750-761, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36367440

RESUMEN

Small GTPases act as molecular switches and control numerous cellular processes by their binding and hydrolysis of guanosine triphosphate (GTP). The activity of small GTPases is coordinated by guanine nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs). Recent structural and functional studies have characterized a subset of GAPs whose catalytic units consist of longin domains. Longin domain containing GAPs regulate small GTPases that facilitate nutrient signalling, autophagy, vesicular trafficking and lysosome homeostasis. All known examples in this GAP family function as part of larger multiprotein complexes. The three characterized mammalian protein complexes in this class are FLCN:FNIP, GATOR1 and C9orf72:SMCR8. Each complex carries out a unique cellular function by regulating distinct small GTPases. In this article, we explore the roles of longin domain GAPs in nutrient sensing, membrane dynamic, vesicular trafficking and disease. Through a structural lens, we examine the mechanism of each longin domain GAP and highlight potential therapeutic applications.


Asunto(s)
Proteínas de Unión al GTP Monoméricas , Transducción de Señal , Animales , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Transporte de Proteínas , Nutrientes , Mamíferos/metabolismo
5.
J Biomol Struct Dyn ; 41(2): 560-580, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-34877916

RESUMEN

Rho family GTPases serve as molecular switches in numerous cellular processes, and their overexpression is involved in disease conditions. RhoG is one of the less explored Rho GTPases with significant sequential and structural homology with Rac1. Experimental mutations in RhoG (i.e., RhoGG12V and RhoGQ61L) are shown to dysregulate cell migration. Thus, targeting upstream activators of RhoG, such as guanine nucleotide exchange factors (GEFs), maybe an important strategy for inhibiting RhoG activation. In the current study, we have modelled the 3D structure of RhoG with greater accuracy as confirmed through PROCHECK, ProSA, and Verify3D. Our results indicate that 90.4% of residues are in the Ramachandran plots favoured region, with the Z-score of -6.46, and 87.96% of residues had an averaged 3D-1D score ≥0.2. Further, we have evaluated and binding dynamics of ten Rac1 inhibitors to investigate their potential to inhibit RhoG by targeting GEFs binding grooves. To this end, the binding energy of the docked complexes of the wild-type (WT) RhoG and its mutant proteins with inhibitor molecules was calculated using the MM/PBSA method. Our results from docking studies showed that macrolide1 binds efficiently with the GEF site of WT RhoG and the mutants mentioned above. However, an extensive analysis using MD simulations (200 ns) showed that the Rac1 based inhibitor, EHop-016, and NSC23766 might bind with greater affinity to GEF sites of mutants and WT RhoG. Thus, the results from the study indicate that Rac1 inhibitors have the potential for use as therapeutics in conditions involving dysregulation of RhoG.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Proteína de Unión al GTP cdc42 , Proteína de Unión al GTP rac1 , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/química , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo , Transducción de Señal , Factores de Intercambio de Guanina Nucleótido/química , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas de Unión al GTP rho
6.
Protoplasma ; 260(4): 1007-1029, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36525153

RESUMEN

Plants are frequently subjected to abiotic and biotic stress which causes major impediments in their growth and development. It is emerging that small guanosine triphosphatases (small GTPases), also known as monomeric GTP-binding proteins, assist plants in managing environmental stress. Small GTPases function as tightly regulated molecular switches that get activated with the aid of guanosine triphosphate (GTP) and deactivated by the subsequent hydrolysis of GTP to guanosine diphosphate (GDP). All small GTPases except Rat sarcoma (Ras) are found in plants, including Ras-like in brain (Rab), Rho of plant (Rop), ADP-ribosylation factor (Arf) and Ras-like nuclear (Ran). The members of small GTPases in plants interact with several downstream effectors to counteract the negative effects of environmental stress and disease-causing pathogens. In this review, we describe processes of stress alleviation by developing pathways involving several small GTPases and their associated proteins which are important for neutralizing fungal infections, stomatal regulation, and activation of abiotic stress-tolerant genes in plants. Previous reviews on small GTPases in plants were primarily focused on Rab GTPases, abiotic stress, and membrane trafficking, whereas this review seeks to improve our understanding of the role of all small GTPases in plants as well as their interactome in regulating mechanisms to combat abiotic and biotic stress. This review brings to the attention of scientists recent research on small GTPases so that they can employ genome editing tools to precisely engineer economically important plants through the overexpression/knock-out/knock-in of stress-related small GTPase genes.


Asunto(s)
Estrés Fisiológico , Proteínas de Unión al GTP rab , Proteínas de Unión al GTP rab/metabolismo , Guanosina Trifosfato/metabolismo
7.
Mol Med Rep ; 27(2)2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36562344

RESUMEN

Ras­related C3 botulinum toxin substrate 1 (RAC1), a member of the Rac family of guanosine triphosphate phosphohydrolases, has been suggested to be a regulator of myocardial injury during ischemia and reperfusion (I/R). Whether microRNAs (miRs) are involved in the regulation of the aforementioned process remains to be elucidated. In the present study, an in vitro model of H9C2 cardiomyocytes was used to establish the overexpression of RAC1 following hypoxia and reoxygenation (H/R). Overexpression of RAC1 in H/R­cultured cardiomyocytes could lead to cellular accumulation of reactive oxygen species (ROS) and facilitate the induction of apoptosis of H9C2 cardiomyocytes during H/R. Subsequent bioinformatic analysis indicated that RAC1 was the target of miRNA­194­5p. Further experiments showed that miR­194­5p attenuated the accumulation of cellular ROS and alleviated the induction of apoptosis of H9C2 cardiomyocytes caused by H/R, which was accompanied by the reduction in the expression levels of the RAC1 protein. Taken together, these results indicated that upregulation of miR­194­5p may function as a self­regulated cardioprotective response against RAC1­mediated ROS accumulation and cardiomyocyte apoptosis. Exogenous administration of miR­194­5p may be a novel target to ameliorate I/R injury­induced myocardial apoptosis.


Asunto(s)
Apoptosis , MicroARNs , Miocitos Cardíacos , Proteína de Unión al GTP rac1 , Humanos , Apoptosis/genética , Hipoxia de la Célula/genética , Línea Celular , Hipoxia/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Miocitos Cardíacos/metabolismo , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Ratas
8.
Cells ; 11(23)2022 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-36496968

RESUMEN

Small GTPases act as molecular switches in regulating a myriad of cellular signaling, cytoskeletal dynamics, vesicular trafficking, and membrane/organelle transport processes. Here, I provide an editorial overview of papers collected in this Special Issue on the "Regulation and Function of Small GTPases 2.0".


Asunto(s)
Proteínas de Unión al GTP Monoméricas , Proteínas de Unión al GTP Monoméricas/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Transducción de Señal , Citoesqueleto/metabolismo
9.
Front Immunol ; 13: 1075386, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36524124

RESUMEN

Phagocytosis, macropinocytosis, and G protein coupled receptor-mediated chemotaxis are Ras-regulated and actin-driven processes. The common regulator for Ras activity in these three processes remains unknown. Here, we show that C2GAP2, a Ras GTPase activating protein, highly expressed in the vegetative growth state in model organism Dictyostelium. C2GAP2 localizes at the leading edge of chemotaxing cells, phagosomes during phagocytosis, and macropinosomes during micropinocytosis. c2gapB- cells lacking C2GAP2 displayed increased Ras activation upon folic acid stimulation and subsequent impaired chemotaxis in the folic acid gradient. In addition, c2gaB- cells have elevated phagocytosis and macropinocytosis, which subsequently results in faster cell growth. C2GAP2 binds multiple phospholipids on the plasma membrane and the membrane recruitment of C2GAP2 requires calcium. Taken together, we show a shared negative regulator of Ras signaling that mediates Ras signaling for chemotaxis, phagocytosis, and macropinocytosis.


Asunto(s)
Dictyostelium , Dictyostelium/metabolismo , Quimiotaxis , Pinocitosis/fisiología , Fagocitosis , Ácido Fólico
10.
Front Immunol ; 13: 1020117, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36341344

RESUMEN

Chemotaxis plays an essential role in recruitment of leukocytes to sites of inflammation. Eukaryotic cells sense chemoattractant with G protein-coupled receptors (GPCRs) and chemotax toward gradients with an enormous concentration range through adaptation. Cells in adaptation no longer respond to the present stimulus but remain sensitive to stronger stimuli. Thus, adaptation provides a fundamental strategy for eukaryotic cells to chemotax through a gradient. Ras activation is the first step in the chemosensing GPCR signaling pathways that displays a transient activation behavior in both model organism Dictyostelium discoideum and mammalian neutrophils. Recently, it has been revealed that C2GAP1 and CAPRI control the GPCR-mediated adaptation in D. discoideum and human neutrophils, respectively. More importantly, both Ras inhibitors regulate the sensitivity of the cells. These findings suggest an evolutionarily conserved molecular mechanism by which eukaryotic cells gate concentration range of chemoattractants for chemotaxis.


Asunto(s)
Quimiotaxis , Dictyostelium , Animales , Humanos , Quimiotaxis/fisiología , Dictyostelium/metabolismo , Factores Quimiotácticos/farmacología , Factores Quimiotácticos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Mamíferos/metabolismo
11.
Hypertension ; 79(12): 2843-2853, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36259376

RESUMEN

BACKGROUND: RGS (regulator of G protein signaling) family members catalyze the termination of G protein signaling cascades. Single nucleotide polymorphisms in the RGS2 gene in humans have been linked to hypertension, preeclampsia, and anxiety disorders. Mice deficient for Rgs2 (Rgs2Null) exhibit hypertension, anxiety, and altered adipose development and function. METHODS: To study cell-specific functions of RGS2, a novel gene-targeted mouse harboring a conditional allele for the Rgs2 gene (Rgs2Flox) was developed. These mice were bred with mice expressing Cre-recombinase via the Agouti-related peptide locus (Agrp-Cre) to cause deletion of Rgs2 from all cells expressing Agrp (Rgs2Agrp-KO), or a novel transgenic mouse expressing Cre-recombinase via the ANG (angiotensin) type 1A receptor (Agtr1a/ AT1A) promoter encoded in a bacterial artificial chromosome (BAC-AT1A-Cre) to delete Rgs2 in all Agtr1a-expressing cells (Rgs2AT1A-KO). RESULTS: Whereas Rgs2Flox, Rgs2Agrp-KO, and BAC-AT1A-Cre mice exhibited normal growth and survival, Rgs2AT1A-KO exhibited pre-weaning lethality. Relative to littermates, Rgs2Agrp-KO exhibited reduced fat gains when maintained on a high fat diet, associated with increased energy expenditure. Similarly, surviving adult Rgs2AT1A-KO mice also exhibited increased energy expenditure. Surprisingly, given the hypertensive phenotype previously reported for Rgs2Null mice and evidence supporting a role for RGS2 in terminating AT1A signaling in various cell types, Rgs2AT1A-KO mice exhibited normal blood pressure, ingestive behaviors, and renal functions, both before and after chronic infusion of ANG (490 ng/kg/min, sc). CONCLUSIONS: These results demonstrate the development of a novel mouse with conditional expression of Rgs2 and illustrate the role of Rgs2 within selected cell types for cardiometabolic control.


Asunto(s)
Hipertensión , Proteínas RGS , Animales , Ratones , Proteína Relacionada con Agouti , Hipertensión/genética , Ratones Noqueados , Ratones Transgénicos , Receptor de Angiotensina Tipo 1/genética , Recombinasas , Proteínas RGS/genética
12.
Traffic ; 23(10): 478-495, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36068165

RESUMEN

Fission yeast cytokinesis is driven by simultaneous septum synthesis, membrane furrowing and actomyosin ring constriction. The septum consists of a primary septum flanked by secondary septa. First, delivery of the glucan synthase Bgs1 and membrane vesicles initiate primary septum synthesis and furrowing. Next, Bgs4 is delivered for secondary septum formation. It is unclear how septum synthesis is coordinated with membrane furrowing. Cdc42 promotes delivery of Bgs1 but not Bgs4. We find that after primary septum initiation, Cdc42 inactivators Rga4 and Rga6 localize to the division site. In rga4Δrga6Δ mutants, Cdc42 activity is enhanced during late cytokinesis and cells take longer to separate. Electron micrographs of the division site in these mutants exhibit malformed septum with irregular membrane structures. These mutants have a larger division plane with enhanced Bgs1 delivery but fail to enhance accumulation of Bgs4 and several exocytic proteins. Additionally, these mutants show endocytic defects at the division site. This suggests that Cdc42 regulates primary septum formation and only certain membrane trafficking events. As cytokinesis progresses Rga4 and Rga6 localize to the division site to decrease Cdc42 activity to allow coupling of Cdc42-independent membrane trafficking events with septum formation for proper septum morphology.


Asunto(s)
Citocinesis , Proteínas Activadoras de GTPasa , Proteínas de Schizosaccharomyces pombe , Actomiosina/metabolismo , Citocinesis/genética , Citocinesis/fisiología , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Schizosaccharomyces , Proteínas de Schizosaccharomyces pombe/metabolismo
13.
Biol Reprod ; 107(2): 619-634, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35403672

RESUMEN

During spermiogenesis, the formation of the mitochondrial sheath is critical for male fertility. The molecular processes that govern the development of the mitochondrial sheath remain unknown. Whether TBC1D21 serves as a GTPase-activating protein (GAP) for GTP hydrolysis in the testis is unclear, despite recent findings indicating that it collaborates with numerous proteins to regulate the formation of the mitochondrial sheath. To thoroughly examine the property of TBC1D21 in spermiogenesis, we applied the CRISPR/Cas9 technology to generate the Tbc1d21-/- mice, Tbc1d21D125A R128K mice with mutation in the GAP catalytic residues (IxxDxxR), and Tbc1d21-3xFlag mice. Male Tbc1d21-/- mice were infertile due to the curved spermatozoa flagella. In vitro fertilization is ineffective for Tbc1d21-/- sperm, although healthy offspring were obtained by intracytoplasmic sperm injection. Electron microscopy revealed aberrant ultrastructural changes in the mitochondrial sheath. Thirty-four Rab vectors were constructed followed by co-immunoprecipitation, which identified RAB13 as a novel TBC1D21 binding protein. Interestingly, infertility was not observed in Tbc1d21D125A R128K mice harboring the catalytic residue, suggesting that TBC1D21 is not a typical GAP for Rab-GTP hydrolysis. Moreover, TBC1D21 was expressed in the sperm mitochondrial sheath in Tbc1d21-3xFlag mice. Immunoprecipitation-mass spectrometry demonstrated the interactions of TBC1D21 with ACTB, TPM3, SPATA19, and VDAC3 to regulate the architecture of the sperm midpiece. The collective findings suggest that TBC1D21 is a scaffold protein required for the organization and stabilization of the mitochondrial sheath morphology.


Asunto(s)
Infertilidad Masculina , Semen , Animales , Proteínas Activadoras de GTPasa/genética , Guanosina Trifosfato/metabolismo , Infertilidad Masculina/genética , Infertilidad Masculina/metabolismo , Masculino , Ratones , Ratones Noqueados , Semen/metabolismo , Cola del Espermatozoide , Espermatogénesis/fisiología , Espermatozoides/metabolismo , Proteínas de Unión al GTP rab/genética
14.
Small GTPases ; 13(1): 239-254, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34565293

RESUMEN

GTP binding proteins known as small GTPases make up one of the largest groups of regulatory proteins and control almost all functions of living cells. Their activity is under, respectively, positive and negative regulation by guanine nucleotide exchange factors (GEFs) and GTPase activating proteins (GAPs), which together with their upstream regulators and the downstream targets of the small GTPases form formidable signalling networks. While genomics has revealed the large size of the GTPase, GEF and GAP repertoires, only a small fraction of their interactions and functions have yet been experimentally explored. Dictyostelid social amoebas have been particularly useful in unravelling the roles of many proteins in the Rac-Rho and Ras-Rap families of GTPases in directional cell migration and regulation of the actin cytoskeleton. Genomes and cell-type specific and developmental transcriptomes are available for Dictyostelium species that span the 0.5 billion years of evolution of the group from their unicellular ancestors. In this work, we identified all GTPases, GEFs and GAPs from genomes representative of the four major taxon groups and investigated their phylogenetic relationships and evolutionary conservation and changes in their functional domain architecture and in their developmental and cell-type specific expression. We performed a hierarchical cluster analysis of the expression profiles of the ~2000 analysed genes to identify putative interacting sets of GTPases, GEFs and GAPs, which highlight sets known to interact experimentally and many novel combinations. This work represents a valuable resource for research into all fields of cellular regulation.


Asunto(s)
Dictyostelium , Proteínas de Unión al GTP Monoméricas , Dictyostelium/genética , Dictyostelium/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Filogenia
15.
Acta Pharm Sin B ; 11(11): 3433-3446, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34900528

RESUMEN

RAS, a member of the small GTPase family, functions as a binary switch by shifting between inactive GDP-loaded and active GTP-loaded state. RAS gain-of-function mutations are one of the leading causes in human oncogenesis, accounting for ∼19% of the global cancer burden. As a well-recognized target in malignancy, RAS has been intensively studied in the past decades. Despite the sustained efforts, many failures occurred in the earlier exploration and resulted in an 'undruggable' feature of RAS proteins. Phosphorylation at several residues has been recently determined as regulators for wild-type and mutated RAS proteins. Therefore, the development of RAS inhibitors directly targeting the RAS mutants or towards upstream regulatory kinases supplies a novel direction for tackling the anti-RAS difficulties. A better understanding of RAS phosphorylation can contribute to future therapeutic strategies. In this review, we comprehensively summarized the current advances in RAS phosphorylation and provided mechanistic insights into the signaling transduction of associated pathways. Importantly, the preclinical and clinical success in developing anti-RAS drugs targeting the upstream kinases and potential directions of harnessing allostery to target RAS phosphorylation sites were also discussed.

16.
Cells ; 10(11)2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34831205

RESUMEN

Rho guanosine triphosphate hydrolases (GTPases) are molecular switches that cycle between an inactive guanosine diphosphate (GDP)-bound and an active guanosine triphosphate (GTP)-bound state during signal transduction. As such, they regulate a wide range of both cellular and physiological processes. In this review, we will summarize recent work on the role of Rho GTPase-regulated pathways in skeletal muscle development, regeneration, tissue mass homeostatic balance, and metabolism. In addition, we will present current evidence that links the dysregulation of these GTPases with diseases caused by skeletal muscle dysfunction. Overall, this information underscores the critical role of a number of members of the Rho GTPase subfamily in muscle development and the overall metabolic balance of mammalian species.


Asunto(s)
Homeostasis , Desarrollo de Músculos , Músculo Esquelético/enzimología , Músculo Esquelético/crecimiento & desarrollo , Proteínas de Unión al GTP rho/metabolismo , Animales , Humanos , Enfermedades Musculares/enzimología , Enfermedades Musculares/patología , Regeneración/fisiología
17.
J Hematol Oncol ; 14(1): 171, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34663417

RESUMEN

GTPase-activating protein (GAP) is a negative regulator of GTPase protein that is thought to promote the conversion of the active GTPase-GTP form to the GTPase-GDP form. Based on its ability to regulate GTPase proteins and other domains, GAPs are directly or indirectly involved in various cell requirement processes. We reviewed the existing evidence of GAPs regulating regulated cell death (RCD), mainly apoptosis and autophagy, as well as some novel RCDs, with particular attention to their association in diseases, especially cancer. We also considered that GAPs could affect tumor immunity and attempted to link GAPs, RCD and tumor immunity. A deeper understanding of the GAPs for regulating these processes could lead to the discovery of new therapeutic targets to avoid pathologic cell loss or to mediate cancer cell death.


Asunto(s)
Proteínas Activadoras de GTPasa/inmunología , Neoplasias/inmunología , Muerte Celular Regulada , Animales , GTP Fosfohidrolasas/inmunología , Guanosina Trifosfato/inmunología , Humanos , Neoplasias/patología
18.
Cell Rep ; 36(9): 109541, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34469730

RESUMEN

The regulation of lipid homeostasis is not well understood. Using forward genetic screening, we demonstrate that the loss of dTBC1D22, an essential gene that encodes a Tre2-Bub2-Cdc16 (TBC) domain-containing protein, results in lipid droplet accumulation in multiple tissues. We observe that dTBC1D22 interacts with Rab40 and exhibits GTPase activating protein (GAP) activity. Overexpression of either the GTP- or GDP-binding-mimic form of Rab40 results in lipid droplet accumulation. We observe that Rab40 mutant flies are defective in lipid mobilization. The lipid depletion induced by overexpression of Brummer, a triglyceride lipase, is dependent on Rab40. Rab40 mutant flies exhibit decreased lipophagy and small size of autolysosomal structures, which may be due to the defective Golgi functions. Finally, we demonstrate that Rab40 physically interacts with Lamp1, and Rab40 is required for the distribution of Lamp1 during starvation. We propose that dTBC1D22 functions as a GAP for Rab40 to regulate lipophagy.


Asunto(s)
Autofagia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Ojo/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Metabolismo de los Lípidos , Proteínas de Unión al GTP rab/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/ultraestructura , Ojo/ultraestructura , Proteínas Activadoras de GTPasa/genética , Aparato de Golgi/genética , Aparato de Golgi/metabolismo , Aparato de Golgi/ultraestructura , Células HeLa , Homeostasis , Humanos , Lipasa/genética , Lipasa/metabolismo , Gotas Lipídicas/metabolismo , Proteína 1 de la Membrana Asociada a los Lisosomas/genética , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Lisosomas/genética , Lisosomas/metabolismo , Lisosomas/ultraestructura , Mutación , Proteínas de Unión al GTP rab/genética
19.
Cytoskeleton (Hoboken) ; 78(3): 67-76, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33792196

RESUMEN

Rho-GTPase-activating proteins (Rho-GAPs) are essential upstream regulators of the Rho family of GTPases. Currently, it remains unclear if the phenotypic change caused by perturbations to a Rho-GAP is predictable from its amino acid sequence. Here we analyze the relationship between the morphological response of cells to the silencing of Rho-GAPs and their primary structure. For all possible pairs of 57 different Rho-GAPs expressed in MCF10A epithelial cells, the similarity in the Rho-GAP silencing-induced morphological change was quantified and compared to the similarity in the primary structure of the corresponding pairs. We found a distinct correlation between the morphological and sequence similarities in a specific group of RhoA-targeting Rho-GAPs. Thus, the family-wide analysis revealed a common feature shared by the specific Rho-GAPs.


Asunto(s)
Células Epiteliales , Proteínas Activadoras de GTPasa , Células Epiteliales/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
20.
Transl Cancer Res ; 10(12): 5085-5094, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35116360

RESUMEN

BACKGROUND: The role of Ral and RalGAPs on the progression of head and neck squamous cell carcinoma (HNSC) remains unclear. METHODS: The predesigned siRNAs against RalGAPs were transfected into cells to evaluate the effect on RalA activation. The Data from TCGA and GTEx were combined to analyze the pan-cancer gene expression of RalA and RalGAPs in cancer and adjacent normal tissues. Kaplan-Meier analysis was used to assess the predictive value of RalA and RalGAPs expression on the overall survival of patients with HNSC. Methylation-specific PCR in vitro and next-generation bisulfite sequencing in vivo were used to evaluate the association between DNA methylation and the down-regulation of RalGAPs. RESULTS: RalGAPs negatively regulated RalA activation. HNSC patients with low level of RalGAPα2 had worse overall survival. The promoter of RalGAPα2 was widely methylated in comparison to RalGAPα1 and the DNA methylation level of RalGAPα2 promoter was increased in HNSC tissues and associated with the presence of neck lymph node metastasis. CONCLUSIONS: RalA and RalGAPs could act as a specific predictor to assess the prognosis of HNSC. DNA methylation might be a potential mechanism that downregulated the RalGAPα2 expression.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA