Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 19(4): e0295103, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38574162

RESUMEN

The ADP-ribosylation factors (Arfs) constitute a family of small GTPases within the Ras superfamily, with a distinguishing structural feature of a hypervariable N-terminal extension of the G domain modified with myristate. Arf proteins, including Arf1, have roles in membrane trafficking and cytoskeletal dynamics. While screening for Arf1:small molecule co-crystals, we serendipitously solved the crystal structure of the non-myristoylated engineered mutation [L8K]Arf1 in complex with a GDP analogue. Like wild-type (WT) non-myristoylated Arf1•GDP, we observed that [L8K]Arf1 exhibited an N-terminal helix that occludes the hydrophobic cavity that is occupied by the myristoyl group in the GDP-bound state of the native protein. However, the helices were offset from one another due to the L8K mutation, with a significant change in position of the hinge region connecting the N-terminus to the G domain. Hypothesizing that the observed effects on behavior of the N-terminus affects interaction with regulatory proteins, we mutated two hydrophobic residues to examine the role of the N-terminal extension for interaction with guanine nucleotide exchange factors (GEFs) and GTPase Activating Proteins (GAPs. Different than previous studies, all mutations were examined in the context of myristoylated Arf. Mutations had little or no effect on spontaneous or GEF-catalyzed guanine nucleotide exchange but did affect interaction with GAPs. [F13A]myrArf1 was less than 1/2500, 1/1500, and 1/200 efficient as substrate for the GAPs ASAP1, ARAP1 and AGAP1; however, [L8A/F13A]myrArf1 was similar to WT myrArf1. Using molecular dynamics simulations, the effect of the mutations on forming alpha helices adjacent to a membrane surface was examined, yet no differences were detected. The results indicate that lipid modifications of GTPases and consequent anchoring to a membrane influences protein function beyond simple membrane localization. Hypothetical mechanisms are discussed.


Asunto(s)
Proteínas Activadoras de GTPasa , Miristatos , Proteínas Activadoras de GTPasa/metabolismo , Mutación Puntual , Ácido Mirístico , Factor 1 de Ribosilacion-ADP/genética , Factor 1 de Ribosilacion-ADP/metabolismo , Factores de Ribosilacion-ADP/genética , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo
2.
J Anim Sci ; 1012023 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-37632755

RESUMEN

Neutering is a significant risk factor for obesity in dogs. Changes in gut microbiota and its metabolites have been identified as a key player during obesity progression. However, the mechanisms that promote neuter-associated weight gain are not well understood. Therefore, in this study, sixteen clinically healthy Beagle dogs (6 male and 10 female, mean age = 8.22 ±â€…0.25 mo old) were neutered. Body weight (BW) and body condition score (BCS) were recorded at 1 d before neutering, 3, 6, 10, 16, and 21 mo after neutering. Dogs were grouped based on their BCS as ideal weight group (IW, n = 4, mean BW = 13.22 ±â€…1.30 kg, mean BCS = 5.00 ±â€…0.41) and obese group (OB, n = 12, mean BW = 18.57 ±â€…1.08 kg, mean BCS = 7.92 ±â€…0.82) at 21 mo after neutering. Serum lipid profile, glucose, and hormones and fecal microbiota and short-chain fatty acids (SCFAs) were measured. Our results showed that OB dogs had greater (P < 0.0001) BW (18.57 vs. 13.22 kg), BCS (7.92 vs. 5.00), and average daily gain (12.27 vs. 5.69 g/d) than IW dogs at 21 mo after neutering, and the obesity rate was up to 60%. In addition, significant increases (P < 0.05) in serum triglyceride (TG, 1.10 vs. 0.56 mmol/L) and high-density lipoprotein cholesterol (HDL-C, 6.96 vs. 5.40 mmol/L) levels and a significant decrease (P < 0.05) in serum adiponectin (APN, 54.06 vs. 58.39 µg/L) level were observed in OB dogs; serum total cholesterol (4.83 vs. 3.75 mmol/L) (P = 0.075) and leptin (LEP, 2.82 vs. 2.53 µg/L) (P = 0.065) levels tended to be greater in OB dogs; there was a trend towards a lower (P = 0.092) APN/LEP (19.32 vs. 21.81) in OB dogs. Results of fecal microbial alpha-diversity showed that Observed_species and Chao1 indices tended to be lower (P = 0.069) in OB dogs. The STAMP and LEfSe analyses revealed that OB dogs had a greater (P < 0.05 and LDA > 2) reduction in relative abundances of Bacteroides, Prevotella_9, and Megamonas than IW dogs. In addition, OB dogs also had greater (P < 0.05) reduction in fecal acetate, propionate, and butyrate concentrations than IW dogs. Moreover, clear negative correlations (|r| > 0.5 and P < 0.05) were found between SCFAs-producing bacteria and BW, TG, and HDL-C. The functional predictions of microbial communities based on PICRUSt2 analysis revealed that lipid metabolism and endocrine system were significantly disturbed in obese dogs after neutering. Thus, intervention with SCFAs-producing bacteria might represent a new target for the prevention or treatment of canine obesity after neutering. Moreover, weight control before neutering may also contribute to the prevention of canine obesity after neutering.


Neutering contributes to canine obesity risk. In this study, obesity rate of 60% at 21 mo after neutering was observed. Obese dogs had greater serum triglyceride, total cholesterol, high-density lipoprotein cholesterol, and leptin levels and lower adiponectin level than ideal weight dogs. In addition, fecal microbiota analysis found a decreasing microbial diversity in obese dogs, and decreasing SCFAs-producing bacteria Megamonas, Bacteroides, and Prevotella_9 in obese dogs resulted in lower production of fecal acetate, propionate, and butyrate. Importantly, strong negative correlations between SCFAs-producing bacteria and body weight, TG, and HDL-C revealed that SCFAs-producing bacteria are involved in the process of canine obesity after neutering. Thus, intervention with SCFAs-producing bacteria may be a target for the prevention or treatment of canine obesity after neutering. Moreover, weight control before neutering may also contribute to the prevention of canine obesity after neutering.


Asunto(s)
Enfermedades de los Perros , Microbioma Gastrointestinal , Perros , Animales , Masculino , Femenino , Obesidad/veterinaria , Obesidad/metabolismo , Ácidos Grasos Volátiles , Factores de Riesgo , Heces/microbiología , Bacterias , Colesterol , Enfermedades de los Perros/microbiología
3.
J Biol Chem ; 299(3): 102992, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36758799

RESUMEN

The ADP-ribosylation factor (Arf) GTPases and their regulatory proteins are implicated in cancer progression. NAV-2729 was previously identified as a specific inhibitor of Arf6 that reduced progression of uveal melanoma in an orthotopic xenograft. Here, our goal was to assess the inhibitory effects of NAV-2729 on the proliferation of additional cell types. We found NAV-2729 inhibited proliferation of multiple cell lines, but Arf6 expression did not correlate with NAV-2729 sensitivity, and knockdown of Arf6 affected neither cell viability nor sensitivity to NAV-2729. Furthermore, binding to native Arf6 was not detected; however, we determined that NAV-2729 inhibited both Arf exchange factors and Arf GTPase-activating proteins. ASAP1, a GTPase-activating protein linked to cancer progression, was further investigated. We demonstrated that NAV-2729 bound to the PH domain of ASAP1 and changed ASAP1 cellular distribution. However, ASAP1 knockdown did not fully recapitulate the cytoskeletal effects of NAV-2729 nor affect cell proliferation. Finally, our screens identified 48 other possible targets of NAV-2729. These results illustrate the complexities of defining targets of small molecules and identify NAV-2729 as a model PH domain-binding inhibitor.


Asunto(s)
Factores de Ribosilacion-ADP , Neoplasias , Humanos , Factores de Ribosilacion-ADP/metabolismo , Clorobencenos , Pirazoles , Proteínas Activadoras de GTPasa/metabolismo , Factor 1 de Ribosilacion-ADP/metabolismo
4.
J Anim Sci ; 1012023 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-36734030

RESUMEN

Inappropriate dietary management may lead to delayed recovery from castration surgery and significant weight gain in cats after castration. Wet canned food often exhibits more advantageous characteristics than dry food (e.g., higher palatability and digestibility, and lower energy density). This study compared the effects of canned and dry food on surgical recovery and weight management in cats after castration. Eighteen healthy cats (weighed 4.33 ± 1.04 kg and aged 18-months old) were allocated to one of the two dietary treatments (N = 9/group), dry (CON) and canned food (CAN) balanced for sex and initial BW. Cats were fed ad libitum for 7 weeks, including one week before surgery (week 0) and 6 weeks after surgery (week 1-6). Daily dry matter intake (DMI), and weekly body weight (BW) and body condition score (BCS) was obtained. Feces were collected for measuring nutrient digestibility and concentrations of short-chain fatty acids (SCFA) and branched-chain fatty acids (BCFA). Physical pain and wound surface assessment were performed at week 1. Blood was also collected intermittently for measuring biochemical indices and untargeted metabolomics analysis. Results indicated that BW, BCS and daily DMI in CON group increased (P < 0.05) over time after castration, but were maintained relatively stable in CAN group. Cats in CAN group exhibited less pain-related behavior as reflected by lower score of comfort (P < 0.05) and vocalization (P < 0.10), improved wound surface assessment (P < 0.10), lower level of lipase (P < 0.10) and ratio of blood urea nitrogen/serum creatinine (BUN/SC; P < 0.05), and higher level of superoxide dismutase (SOD; P < 0.05) in week 1 than CON cats. Meanwhile, the CAN group had significantly higher concentration of immunoglobulin G (IgG) on days 5 and 7, and higher level of high-density lipoprotein cholesterol (HDL-C; P < 0.10) but lower triglyceride (TG; P < 0.05) than CON group on day 20 and 48. Fecal total and most individual SCFA increased significantly from week 1 to week 6 regardless of diet, but the increase of butyric acid over time only occurred in CON group (P < 0.05). Also, serum metabolomic analysis revealed differential metabolic pathways between the two groups. Overall, compared with the dry food, the canned food tested in our study promoted cat wound recovery by reducing pain and increasing immune and antioxidative capacity after sterilizing surgery, and helped to maintain healthy body condition in cats after castration.


Castration is a surgical operation common in pet cats and dogs, and weight gain is often observed a period after castration. Nutritional management can be important for animal health in both processes. Due to differences in manufacturing techniques and nutrient composition, wet canned food generally exhibits higher palatability and lower energy density than dry food. Till date, few studies have explored if compared to dry kibbles, canned diet can have advantages in promoting recovery from castration surgery and maintaining normal body condition after castration in cats. In our study, dry and canned diets were fed to cats experiencing castration surgery with a free-feeding method. During the one week after surgery, cats fed canned food exhibited less pain and discomfort, and improved inflammation and antioxidative capacity than cats fed dry food. During the four weeks after surgery, cats fed dry food showed significantly more weight gain and change of body condition, meanwhile their blood and fecal measures resembled more of those observed in overweight and/or obese individuals than cats fed canned food. Collectively, canned food with high palatability and low energy density promoted the recovery of cats from the castration surgery and reduced their weight gain after castration.


Asunto(s)
Dieta , Ácidos Grasos , Masculino , Gatos , Animales , Peso Corporal , Dieta/veterinaria , Heces/química , Ácidos Grasos/análisis , Ácidos Grasos Volátiles , Orquiectomía/veterinaria , Alimentación Animal/análisis , Digestión
5.
Biochim Biophys Acta Mol Cell Res ; 1869(8): 119264, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35381293

RESUMEN

Osteosarcoma (OS) and Pax-Foxo1 fusion negative rhabdomyosarcoma (FN-RMS) are pediatric sarcomas with poor prognoses in patients with advanced disease. In both malignancies, an actin binding protein has been linked to poor prognosis. Integrin adhesion complexes (IACs) are closely coupled to actin networks and IAC-mediated signaling has been implicated in the progression of carcinomas. However, the relationship of IACs and actin cytoskeleton remodeling with cell signaling is understudied in pediatric sarcomas. Here, we tested the hypothesis that IAC dynamics affect ERK activation in OS and FN-RMS cell lines. Adhesion dependence of ERK activation differed among the OS and FN-RMS cells examined. In the OS cell lines, adhesion did not have a consistent effect on phospho-ERK (pERK). ERK phosphorylation in response to fetal calf serum or 1 ng/ml EGF was nearly as efficient in OS cell lines and one FN-RMS cell line in suspension as cells adherent to poly-l-lysine (PL) or fibronectin (FN). By contrast, adhesion to plastic, PL or FN increased ERK phosphorylation and was greater than additive with a 15 min exposure to 1 ng/ml EGF in three FN-RMS cell lines. Increases in pERK were partly dependent on FAK and PAK1/2 but independent of IAC maturation. As far as we are aware, this examination of adhesion-dependent signaling is the first in pediatric sarcomas and has led to the discovery of differences from the prevailing paradigms and differences in the degree of coupling between components in the signaling pathways among the cell lines.


Asunto(s)
Factor de Crecimiento Epidérmico , Sarcoma , Adhesión Celular , Línea Celular , Niño , Quinasa 1 de Adhesión Focal , Proteína-Tirosina Quinasas de Adhesión Focal , Humanos , Fosforilación , Sarcoma/genética
6.
J Biol Chem ; 298(3): 101700, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35143843

RESUMEN

Actin filament maintenance is critical for both normal cell homeostasis and events associated with malignant transformation. The ADP-ribosylation factor GTPase-activating protein ASAP1 regulates the dynamics of filamentous actin-based structures, including stress fibers, focal adhesions, and circular dorsal ruffles. Here, we have examined the molecular basis for ASAP1 association with actin. Using a combination of structural modeling, mutagenesis, and in vitro and cell-based assays, we identify a putative-binding interface between the N-Bin-Amphiphysin-Rvs (BAR) domain of ASAP1 and actin filaments. We found that neutralization of charges and charge reversal at positions 75, 76, and 79 of ASAP1 reduced the binding of ASAP1 BAR-pleckstrin homology tandem to actin filaments and abrogated actin bundle formation in vitro. In addition, overexpression of actin-binding defective ASAP1 BAR-pleckstrin homology [K75, K76, K79] mutants prevented cellular actin remodeling in U2OS cells. Exogenous expression of [K75E, K76E, K79E] mutant of full-length ASAP1 did not rescue the reduction of cellular actin fibers consequent to knockdown of endogenous ASAP1. Taken together, our results support the hypothesis that the lysine-rich cluster in the N-BAR domain of ASAP1 is important for regulating actin filament organization.


Asunto(s)
Citoesqueleto de Actina , Actinas , Proteínas Adaptadoras Transductoras de Señales , Proteínas Activadoras de GTPasa , Factores de Ribosilacion-ADP/metabolismo , Citoesqueleto de Actina/genética , Citoesqueleto de Actina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Lisina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Unión Proteica , Dominios Proteicos
7.
Nat Commun ; 12(1): 3906, 2021 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-34162842

RESUMEN

Age-related macular degeneration (AMD) is a multifactorial neurodegenerative disorder. Although molecular mechanisms remain elusive, deficits in autophagy have been associated with AMD. Here we show that deficiency of calcium and integrin binding protein 2 (CIB2) in mice, leads to age-related pathologies, including sub-retinal pigment epithelium (RPE) deposits, marked accumulation of drusen markers APOE, C3, Aß, and esterified cholesterol, and impaired visual function, which can be rescued using exogenous retinoids. Cib2 mutant mice exhibit reduced lysosomal capacity and autophagic clearance, and increased mTORC1 signaling-a negative regulator of autophagy. We observe concordant molecular deficits in dry-AMD RPE/choroid post-mortem human tissues. Mechanistically, CIB2 negatively regulates mTORC1 by preferentially binding to 'nucleotide empty' or inactive GDP-loaded Rheb. Upregulated mTORC1 signaling has been implicated in lymphangioleiomyomatosis (LAM) cancer. Over-expressing CIB2 in LAM patient-derived fibroblasts downregulates hyperactive mTORC1 signaling. Thus, our findings have significant implications for treatment of AMD and other mTORC1 hyperactivity-associated disorders.


Asunto(s)
Autofagia/genética , Proteínas de Unión al Calcio/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Epitelio Pigmentado de la Retina/metabolismo , Transducción de Señal/genética , Animales , Células COS , Proteínas de Unión al Calcio/deficiencia , Células Cultivadas , Chlorocebus aethiops , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Lisosomas/metabolismo , Degeneración Macular/genética , Degeneración Macular/patología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Noqueados , Retina/metabolismo
8.
Sci Adv ; 6(40)2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32998886

RESUMEN

Adenosine diphosphate-ribosylation factor (Arf) guanosine triphosphatase-activating proteins (GAPs) are enzymes that need to bind to membranes to catalyze the hydrolysis of guanosine triphosphate (GTP) bound to the small GTP-binding protein Arf. Binding of the pleckstrin homology (PH) domain of the ArfGAP With SH3 domain, ankyrin repeat and PH domain 1 (ASAP1) to membranes containing phosphatidylinositol 4,5-bisphosphate [PI(4,5)P2] is key for maximum GTP hydrolysis but not fully understood. By combining nuclear magnetic resonance, neutron reflectometry, and molecular dynamics simulation, we show that binding of multiple PI(4,5)P2 molecules to the ASAP1 PH domain (i) triggers a functionally relevant allosteric conformational switch and (ii) maintains the PH domain in a well-defined orientation, allowing critical contacts with an Arf1 mimic to occur. Our model provides a framework to understand how binding of the ASAP1 PH domain to PI(4,5)P2 at the membrane may play a role in the regulation of ASAP1.

9.
J Immunol ; 204(4): 1012-1021, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31924649

RESUMEN

Cell polarization is a key step for leukocytes adhesion and transmigration during leukocytes' inflammatory infiltration. Polarized localization of plasma membrane (PM) phosphatidylinositol-4-phosphate (PtdIns4P) directs the polarization of RPH3A, which contains a PtdIns4P binding site. Consequently, RPH3A mediates the RAB21 and PIP5K1C90 polarization, which is important for neutrophil adhesion to endothelia during inflammation. However, the mechanism by which RPH3A is recruited only to PM PtdIns4P rather than Golgi PtdIns4P remains unclear. By using ADP-ribosylation factor 6 (ARF6) small interfering RNA, ARF6 dominant-negative mutant ARF6(T27N), and ARF6 activation inhibitor SecinH3, we demonstrate that ARF6 plays an important role in the polarization of RPH3A, RAB21, and PIP5K1C90 in murine neutrophils. PM ARF6 is polarized and colocalized with RPH3A, RAB21, PIP5K1C90, and PM PtdIns4P in mouse and human neutrophils upon integrin stimulation. Additionally, ARF6 binds to RPH3A and enhances the interaction between the PM PtdIns4P and RPH3A. Consistent with functional roles of polarization of RPH3A, Rab21, and PIP5K1C90, ARF6 is also required for neutrophil adhesion on the inflamed endothelial layer. Our study reveals a previously unknown role of ARF6 in neutrophil polarization as being the coincidence-detection code with PM PtdIns4P. Cooperation of ARF6 and PM PtdIns4P direct RPH3A polarization, which is important for neutrophil firm adhesion to endothelia.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Endotelio/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Activación Neutrófila , Neutrófilos/inmunología , Proteínas de Transporte Vesicular/metabolismo , Factor 6 de Ribosilación del ADP , Animales , Adhesión Celular/inmunología , Línea Celular , Membrana Celular/metabolismo , Movimiento Celular/inmunología , Células Endoteliales , Endotelio/citología , Endotelio/inmunología , Femenino , Voluntarios Sanos , Humanos , Ratones , Neutrófilos/citología , Neutrófilos/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Cultivo Primario de Células , Rabfilina-3A
10.
iScience ; 22: 166-180, 2019 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-31785555

RESUMEN

ASAP1 is a multi-domain ArfGAP that controls cell migration, spreading, and focal adhesion dynamics. Although its GAP activity contributes to remodeling of the actin cytoskeleton, it does not fully explain all cellular functions of ASAP1. Here we find that ASAP1 regulates actin filament assembly directly through its N-BAR domain and controls stress fiber maintenance. ASAP1 depletion caused defects in stress fiber organization. Conversely, overexpression of ASAP1 enhanced actin remodeling. The BAR-PH fragment was sufficient to affect actin. ASAP1 with the BAR domain replaced with the BAR domain of the related ACAP1 did not affect actin. The BAR-PH tandem of ASAP1 bound and bundled actin filaments directly, whereas the presence of the ArfGAP and the C-terminal linker/SH3 domain reduced binding and bundling of filaments by BAR-PH. Together these data provide evidence that ASAP1 may regulate the actin cytoskeleton through direct interaction of the BAR-PH domain with actin filaments.

11.
J Biol Chem ; 294(46): 17354-17370, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31591270

RESUMEN

Arf GAP with Src homology 3 domain, ankyrin repeat, and pleckstrin homology (PH) domain 1 (ASAP1) is a multidomain GTPase-activating protein (GAP) for ADP-ribosylation factor (ARF)-type GTPases. ASAP1 affects integrin adhesions, the actin cytoskeleton, and invasion and metastasis of cancer cells. ASAP1's cellular function depends on its highly-regulated and robust ARF GAP activity, requiring both the PH and the ARF GAP domains of ASAP1, and is modulated by phosphatidylinositol 4,5-bisphosphate (PIP2). The mechanistic basis of PIP2-stimulated GAP activity is incompletely understood. Here, we investigated whether PIP2 controls binding of the N-terminal extension of ARF1 to ASAP1's PH domain and thereby regulates its GAP activity. Using [Δ17]ARF1, lacking the N terminus, we found that PIP2 has little effect on ASAP1's activity. A soluble PIP2 analog, dioctanoyl-PIP2 (diC8PIP2), stimulated GAP activity on an N terminus-containing variant, [L8K]ARF1, but only marginally affected activity on [Δ17]ARF1. A peptide comprising residues 2-17 of ARF1 ([2-17]ARF1) inhibited GAP activity, and PIP2-dependently bound to a protein containing the PH domain and a 17-amino acid-long interdomain linker immediately N-terminal to the first ß-strand of the PH domain. Point mutations in either the linker or the C-terminal α-helix of the PH domain decreased [2-17]ARF1 binding and GAP activity. Mutations that reduced ARF1 N-terminal binding to the PH domain also reduced the effect of ASAP1 on cellular actin remodeling. Mutations in the ARF N terminus that reduced binding also reduced GAP activity. We conclude that PIP2 regulates binding of ASAP1's PH domain to the ARF1 N terminus, which may partially regulate GAP activity.


Asunto(s)
Factor 1 de Ribosilacion-ADP/genética , Factores de Ribosilacion-ADP/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Fosfatidilinositol 4,5-Difosfato/genética , Factor 1 de Ribosilacion-ADP/química , Factores de Ribosilacion-ADP/química , Actinas/química , Actinas/genética , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Activadoras de GTPasa/química , Proteínas Activadoras de GTPasa/genética , Humanos , Neoplasias/genética , Fosfatidilinositol 4,5-Difosfato/química , Dominios Homólogos a Pleckstrina/genética , Mutación Puntual/genética , Unión Proteica/genética
12.
Biol Cell ; 110(12): 257-270, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30144359

RESUMEN

BACKGROUND INFORMATION: ARAP2, an Arf GTPase-activating protein (Arf GAP) that binds to adaptor protein with PH domain, PTB domain and leucine zipper motifs 1 (APPL1), regulates focal adhesions (FAs). APPL1 affects FA dynamics by regulating Akt. Here, we tested the hypothesis that ARAP2 affects FAs in part by regulating Akt through APPL1. RESULTS: We found that ARAP2 controlled FA dynamics dependent on its enzymatic Arf GAP activity. In some cells, ARAP2 also regulated phosphoAkt (pAkt) levels. However, ARAP2 control of FAs did not require Akt and conversely, the effects on pAkt were independent of FAs. Reducing ARAP2 expression reduced the size and number of FAs in U118, HeLa and MDA-MB-231 cells. Decreasing ARAP2 expression increased pAkt in U118 cells and HeLa cells and overexpressing ARAP2 decreased pAkt in U118 cells; in contrast, ARAP2 had no effect on pAkt in MDA-MB-231 cells. An Akt inhibitor did not block the effect of reduced ARAP2 on FAs in U118. Furthermore, the effect of ARAP2 on Akt did not require Arf GAP activity, which is necessary for effects on FAs and integrin traffic. Altering FAs by other means did not induce the same changes in pAkt as those seen by reducing ARAP2 in U118 cells. In addition, we discovered that ARAP2 and APPL1 had co-ordinated effects on pAkt in U118 cells. Reduced APPL1 expression, as for ARAP2, increased pAkt in U118 and the effect of reduced APPL1 expression was reversed by overexpressing ARAP2. Conversely, the effect of reduced ARAP2 expression was reversed by overexpressing APPL1. ARAP2 is an Arf GAP that has previously been reported to affect FAs by regulating Arf6 and integrin trafficking and to bind to the adaptor proteins APPL1. Here, we report that ARAP2 suppresses pAkt levels in cells co-ordinately with APPL1 and independently of GAP activity and its effect on the dynamic behaviour of FAs. CONCLUSIONS: We conclude that ARAP2 affects Akt signalling in some cells by a mechanism independent of FAs or membrane traffic. SIGNIFICANCE: Our results highlight an Arf GAP-independent function of ARAP2 in regulating Akt activity and distinguish the effect of ARAP2 on Akt from that on FAs and integrin trafficking, which requires regulation of Arf6.


Asunto(s)
Proteínas Portadoras/metabolismo , Adhesiones Focales/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Citoplasma/metabolismo , Células HeLa , Humanos , Paxillin/metabolismo , Fosforilación
14.
J Biol Chem ; 291(41): 21350-21362, 2016 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-27531749

RESUMEN

The molecular basis for control of the cytoskeleton by the Arf GTPase-activating protein AGAP1 has not been characterized. AGAP1 is composed of G-protein-like (GLD), pleckstrin homology (PH), Arf GAP, and ankyrin repeat domains. Kif2A was identified in screens for proteins that bind to AGAP1. The GLD and PH domains of AGAP1 bound the motor domain of Kif2A. Kif2A increased GAP activity of AGAP1, and a protein composed of the GLD and PH domains of AGAP1 increased ATPase activity of Kif2A. Knockdown (KD) of Kif2A or AGAP1 slowed cell migration and accelerated cell spreading. The effect of Kif2A KD on spreading could be rescued by expression of Kif2A-GFP or FLAG-AGAP1, but not by Kif2C-GFP. The effect of AGAP1 KD could be rescued by FLAG-AGAP1, but not by an AGAP1 mutant that did not bind Kif2A efficiently, ArfGAP1-HA or Kif2A-GFP. Taken together, the results support the hypothesis that the Kif2A·AGAP1 complex contributes to control of cytoskeleton remodeling involved in cell movement.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Cinesinas/metabolismo , Animales , Bovinos , Células Cultivadas , Proteínas Activadoras de GTPasa/química , Proteínas Activadoras de GTPasa/genética , Células HeLa , Humanos , Cinesinas/química , Cinesinas/genética
15.
J Biol Chem ; 291(14): 7517-26, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26893376

RESUMEN

ASAP1 regulates F-actin-based structures and functions, including focal adhesions (FAs) and circular dorsal ruffles (CDRs), cell spreading and migration. ASAP1 function requires its N-terminal BAR domain. We discovered that nonmuscle myosin 2A (NM2A) directly bound the BAR-PH tandem of ASAP1in vitro ASAP1 and NM2A co-immunoprecipitated and colocalized in cells. Knockdown of ASAP1 reduced colocalization of NM2A and F-actin in cells. Knockdown of ASAP1 or NM2A recapitulated each other's effects on FAs, cell migration, cell spreading, and CDRs. The NM2A-interacting BAR domain contributed to ASAP1 control of cell spreading and CDRs. Exogenous expression of NM2A rescued the effect of ASAP1 knockdown on CDRs but ASAP1 did not rescue NM2A knockdown defect in CDRs. Our results support the hypothesis that ASAP1 is a positive regulator of NM2A. Given other binding partners of ASAP1, ASAP1 may directly link signaling and the mechanical machinery of cell migration.


Asunto(s)
Actomiosina/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Movimiento Celular/fisiología , Miosina Tipo IIA no Muscular/metabolismo , Transducción de Señal/fisiología , Actomiosina/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Células HeLa , Humanos , Ratones , Células 3T3 NIH , Miosina Tipo IIA no Muscular/genética , Unión Proteica , Estructura Terciaria de Proteína
16.
Structure ; 23(11): 1977-88, 2015 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-26365802

RESUMEN

We have defined the molecular basis for association of the PH domain of the Arf GAP ASAP1 with phospholipid bilayers. Structures of the unliganded and dibutyryl PtdIns(4,5)P2-bound PH domain were solved. PtdIns(4,5)P2 made contact with both a canonical site (C site) and an atypical site (A site). We hypothesized cooperative binding of PtdIns(4,5)P2 to the C site and a nonspecific anionic phospholipid to the A site. PtdIns(4,5)P2 dependence of binding to large unilamellar vesicles and GAP activity was sigmoidal, consistent with cooperative sites. In contrast, PtdIns(4,5)P2 binding to the PH domain of PLC δ1 was hyperbolic. Mutation of amino acids in either the C or A site resulted in decreased PtdIns(4,5)P2-dependent binding to vesicles and decreased GAP activity. The results support the idea of cooperative phospholipid binding to the C and A sites of the PH domain of ASAP1. We propose that the mechanism underlies rapid switching between active and inactive ASAP1.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Fosfatidilinositol 4,5-Difosfato/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Humanos , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Fosfatidilinositol 4,5-Difosfato/química , Unión Proteica
17.
Methods Cell Biol ; 130: 69-80, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26360029

RESUMEN

Defining the interaction of Arf GAPs with specific Arfs is important for understanding their functions in the endocytic system. Cell-based approaches have been valuable for identifying Arfs and Arf GAPs active in the endocytic compartment; however, the cell-based assays have some limitations in establishing relationships among the Arfs and ArfGAPs. Here we describe a simple in vitro assay that will provide a means for comparing Arfs as substrates and serve to complement cell-based studies.


Asunto(s)
Factor 1 de Ribosilacion-ADP/química , Factor 1 de Ribosilacion-ADP/biosíntesis , Factor 1 de Ribosilacion-ADP/aislamiento & purificación , Pruebas de Enzimas , Escherichia coli , Guanosina Trifosfato/química , Humanos , Hidrólisis , Liposomas Unilamelares
18.
J Neurosci ; 35(24): 9088-105, 2015 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-26085633

RESUMEN

Mutant genes that underlie Mendelian forms of amyotrophic lateral sclerosis (ALS) and biochemical investigations of genetic disease models point to potential driver pathophysiological events involving endoplasmic reticulum (ER) stress and autophagy. Several steps in these cell biological processes are known to be controlled physiologically by small ADP-ribosylation factor (ARF) signaling. Here, we investigated the role of ARF guanine nucleotide exchange factors (GEFs), cytohesins, in models of ALS. Genetic or pharmacological inhibition of cytohesins protects motor neurons in vitro from proteotoxic insults and rescues locomotor defects in a Caenorhabditis elegans model of disease. Cytohesins form a complex with mutant superoxide dismutase 1 (SOD1), a known cause of familial ALS, but this is not associated with a change in GEF activity or ARF activation. ER stress evoked by mutant SOD1 expression is alleviated by antagonism of cytohesin activity. In the setting of mutant SOD1 toxicity, inhibition of cytohesin activity enhances autophagic flux and reduces the burden of misfolded SOD1. These observations suggest that targeting cytohesins may have potential benefits for the treatment of ALS.


Asunto(s)
Proteínas de Caenorhabditis elegans/antagonistas & inhibidores , Proteínas de Caenorhabditis elegans/genética , Modelos Animales de Enfermedad , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/genética , Enfermedad de la Neurona Motora/genética , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Caenorhabditis elegans , Proteínas de Caenorhabditis elegans/biosíntesis , Células Cultivadas , Proteínas Activadoras de GTPasa/antagonistas & inhibidores , Proteínas Activadoras de GTPasa/biosíntesis , Proteínas Activadoras de GTPasa/genética , Factores de Intercambio de Guanina Nucleótido/biosíntesis , Células HeLa , Humanos , Ratones , Modelos Genéticos , Ratas , Ratas Sprague-Dawley , Superóxido Dismutasa/biosíntesis , Superóxido Dismutasa/genética
19.
J Biol Chem ; 289(44): 30237-30248, 2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25225293

RESUMEN

Arf6 and the Arf6 GTPase-activating protein (GAP) ACAP1 are established regulators of integrin traffic important to cell adhesion and migration. However, the function of Arf6 with ACAP1 cannot explain the range of Arf6 effects on integrin-based structures. We propose that Arf6 has different functions determined, in part, by the associated Arf GAP. We tested this idea by comparing the Arf6 GAPs ARAP2 and ACAP1. We found that ARAP2 and ACAP1 had opposing effects on apparent integrin ß1 internalization. ARAP2 knockdown slowed, whereas ACAP1 knockdown accelerated, integrin ß1 internalization. Integrin ß1 association with adaptor protein containing a pleckstrin homology (PH) domain, phosphotyrosine-binding (PTB) domain, and leucine zipper motif (APPL)-positive endosomes and EEA1-positive endosomes was affected by ARAP2 knockdown and depended on ARAP2 GAP activity. ARAP2 formed a complex with APPL1 and colocalized with Arf6 and APPL in a compartment distinct from the Arf6/ACAP1 tubular recycling endosome. In addition, although ACAP1 and ARAP2 each colocalized with Arf6, they did not colocalize with each other and had opposing effects on focal adhesions (FAs). ARAP2 overexpression promoted large FAs, but ACAP1 overexpression reduced FAs. Taken together, the data support a model in which Arf6 has at least two sites of opposing action defined by distinct Arf6 GAPs.


Asunto(s)
Proteínas Portadoras/fisiología , Endosomas/metabolismo , Proteínas Activadoras de GTPasa/fisiología , Receptores de Vitronectina/metabolismo , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Endocitosis , Receptores ErbB/metabolismo , Adhesiones Focales/metabolismo , Células HeLa , Humanos , Transporte de Proteínas , Transferrina/metabolismo , Proteínas de Transporte Vesicular/metabolismo
20.
J Biol Chem ; 288(8): 5896-913, 2013 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-23288846

RESUMEN

Previously, we reported an acidification-dependent interaction of the endosomal vacuolar H(+)-ATPase (V-ATPase) with cytohesin-2, a GDP/GTP exchange factor (GEF), suggesting that it functions as a pH-sensing receptor. Here, we have studied the molecular mechanism of signaling between the V-ATPase, cytohesin-2, and Arf GTP-binding proteins. We found that part of the N-terminal cytosolic tail of the V-ATPase a2-subunit (a2N), corresponding to its first 17 amino acids (a2N(1-17)), potently modulates the enzymatic GDP/GTP exchange activity of cytohesin-2. Moreover, this peptide strongly inhibits GEF activity via direct interaction with the Sec7 domain of cytohesin-2. The structure of a2N(1-17) and its amino acids Phe(5), Met(10), and Gln(14) involved in interaction with Sec7 domain were determined by NMR spectroscopy analysis. In silico docking experiments revealed that part of the V-ATPase formed by its a2N(1-17) epitope competes with the switch 2 region of Arf1 and Arf6 for binding to the Sec7 domain of cytohesin-2. The amino acid sequence alignment and GEF activity studies also uncovered the conserved character of signaling between all four (a1-a4) a-subunit isoforms of mammalian V-ATPase and cytohesin-2. Moreover, the conserved character of this phenomenon was also confirmed in experiments showing binding of mammalian cytohesin-2 to the intact yeast V-ATPase holo-complex. Thus, here we have uncovered an evolutionarily conserved function of the V-ATPase as a novel cytohesin-signaling receptor.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo , Factores de Ribosilacion-ADP/metabolismo , Secuencia de Aminoácidos , Animales , Dicroismo Circular , ADN Complementario/metabolismo , Epítopos/química , Proteínas de Unión al GTP/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Espectroscopía de Resonancia Magnética/métodos , Ratones , Microscopía Confocal/métodos , Datos de Secuencia Molecular , Péptidos/química , Isoformas de Proteínas , Estructura Secundaria de Proteína , Ratas , Proteínas Recombinantes/química , Transducción de Señal , Triptófano/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...