Protein factors that inhibit the dissociation of GDP from GTP-BINDING PROTEINS.
A subcategory of guanine nucleotide dissociation inhibitors that are specific for RHO GTP-BINDING PROTEINS.
An abundantly-expressed rho GDP-dissociation inhibitor subtype that regulates a broad variety of RHO GTPASES.
A rho GDP-dissociation inhibitor subtype that has a unique C-terminal alpha helical membrane-binding domain. It is found bound to CYTOPLASMIC VESICLES such as those associated with the GOLGI APPARATUS.
A rho GDP-dissociation inhibitor subtype that is highly expressed in hematopoietic cells and in LYMPHOCYTES. The expression of this subtype is associated with the regulation of CELL PROLIFERATION; TUMORIGENESIS; and APOPTOSIS.
A guanine nucleotide containing two phosphate groups esterified to the sugar moiety.
Guanosine 5'-(tetrahydrogen triphosphate). A guanine nucleotide containing three phosphate groups esterified to the sugar moiety.
A large family of MONOMERIC GTP-BINDING PROTEINS that are involved in regulation of actin organization, gene expression and cell cycle progression. This enzyme was formerly listed as EC 3.6.1.47.
A member of the Rho family of MONOMERIC GTP-BINDING PROTEINS. It is associated with a diverse array of cellular functions including cytoskeletal changes, filopodia formation and transport through the GOLGI APPARATUS. This enzyme was formerly listed as EC 3.6.1.47.
A family of heterotrimeric GTP-binding protein alpha subunits that were originally identified by their ability to inhibit ADENYLYL CYCLASES. Members of this family can couple to beta and gamma G-protein subunits that activate POTASSIUM CHANNELS. The Gi-Go part of the name is also spelled Gi/Go.
Regulatory proteins that act as molecular switches. They control a wide range of biological processes including: receptor signaling, intracellular signal transduction pathways, and protein synthesis. Their activity is regulated by factors that control their ability to bind to and hydrolyze GTP to GDP. EC 3.6.1.-.
A guanine nucleotide exchange factor that stimulates the dissociation of GDP from RAL GTP-BINDING PROTEINS. It also has GDP exchange activity towards other MONOMERIC GTP-BINDING PROTEINS.
A RHO GTP-BINDING PROTEIN involved in regulating signal transduction pathways that control assembly of focal adhesions and actin stress fibers. This enzyme was formerly listed as EC 3.6.1.47.
Protein factors that promote the exchange of GTP for GDP bound to GTP-BINDING PROTEINS.
A family of MONOMERIC GTP-BINDING PROTEINS that are related to RAS PROTEINS.This enzyme was formerly listed as EC 3.6.1.47.
A family of ubiquitously expressed MONOMERIC GTP-BINDING PROTEINS that are involved in intracellular signal transduction. This enzyme was formerly listed as EC 3.6.1.47.
Guanine nucleotides are cyclic or linear molecules that consist of a guanine base, a pentose sugar (ribose in the cyclic form, deoxyribose in the linear form), and one or more phosphate groups, playing crucial roles in signal transduction, protein synthesis, and regulation of enzymatic activities.
The process in which substances, either endogenous or exogenous, bind to proteins, peptides, enzymes, protein precursors, or allied compounds. Specific protein-binding measures are often used as assays in diagnostic assessments.
Enzymes that hydrolyze GTP to GDP. EC 3.6.1.-.
The order of amino acids as they occur in a polypeptide chain. This is referred to as the primary structure of proteins. It is of fundamental importance in determining PROTEIN CONFORMATION.
Descriptions of specific amino acid, carbohydrate, or nucleotide sequences which have appeared in the published literature and/or are deposited in and maintained by databanks such as GENBANK, European Molecular Biology Laboratory (EMBL), National Biomedical Research Foundation (NBRF), or other sequence repositories.
Guanosine 5'-(trihydrogen diphosphate), monoanhydride with phosphorothioic acid. A stable GTP analog which enjoys a variety of physiological actions such as stimulation of guanine nucleotide-binding proteins, phosphoinositide hydrolysis, cyclic AMP accumulation, and activation of specific proto-oncogenes.
Guanine is a purine nucleobase, one of the four nucleobases in the nucleic acid of DNA and RNA, involved in forming hydrogen bonds between complementary base pairs in double-stranded DNA molecules.
Signaling proteins which function as master molecular switches by activating Rho GTPases through conversion of guanine nucleotides. Rho GTPases in turn control many aspects of cell behavior through the regulation of multiple downstream signal transduction pathways.
A genetically related subfamily of RAB GTP-BINDING PROTEINS involved in calcium-dependent EXOCYTOSIS. This enzyme was formerly listed as EC 3.6.1.47.
The monomeric units from which DNA or RNA polymers are constructed. They consist of a purine or pyrimidine base, a pentose sugar, and a phosphate group. (From King & Stansfield, A Dictionary of Genetics, 4th ed)
A large family of MONOMERIC GTP-BINDING PROTEINS that play a key role in cellular secretory and endocytic pathways. EC 3.6.1.-.
A family of GUANINE NUCLEOTIDE EXCHANGE FACTORS that are specific for RAS PROTEINS.
A rac GTP-binding protein involved in regulating actin filaments at the plasma membrane. It controls the development of filopodia and lamellipodia in cells and thereby influences cellular motility and adhesion. It is also involved in activation of NADPH OXIDASE. This enzyme was formerly listed as EC 3.6.1.47.
The rate dynamics in chemical or physical systems.

Vac1p coordinates Rab and phosphatidylinositol 3-kinase signaling in Vps45p-dependent vesicle docking/fusion at the endosome. (1/388)

The vacuolar protein sorting (VPS) pathway of Saccharomyces cerevisiae mediates transport of vacuolar protein precursors from the late Golgi to the lysosome-like vacuole. Sorting of some vacuolar proteins occurs via a prevacuolar endosomal compartment and mutations in a subset of VPS genes (the class D VPS genes) interfere with the Golgi-to-endosome transport step. Several of the encoded proteins, including Pep12p/Vps6p (an endosomal target (t) SNARE) and Vps45p (a Sec1p homologue), bind each other directly [1]. Another of these proteins, Vac1p/Pep7p/Vps19p, associates with Pep12p and binds phosphatidylinositol 3-phosphate (PI(3)P), the product of the Vps34 phosphatidylinositol 3-kinase (PI 3-kinase) [1] [2]. Here, we demonstrate that Vac1p genetically and physically interacts with the activated, GTP-bound form of Vps21p, a Rab GTPase that functions in Golgi-to-endosome transport, and with Vps45p. These results implicate Vac1p as an effector of Vps21p and as a novel Sec1p-family-binding protein. We suggest that Vac1p functions as a multivalent adaptor protein that ensures the high fidelity of vesicle docking and fusion by integrating both phosphoinositide (Vps34p) and GTPase (Vps21p) signals, which are essential for Pep12p- and Vps45p-dependent targeting of Golgi-derived vesicles to the prevacuolar endosome.  (+info)

Rho family small G proteins play critical roles in mechanical stress-induced hypertrophic responses in cardiac myocytes. (2/388)

-Mechanical stress induces a variety of hypertrophic responses, such as activation of protein kinases, reprogramming of gene expression, and an increase in protein synthesis. In the present study, to elucidate how mechanical stress induces such events, we examined the role of Rho family small GTP-binding proteins (G proteins) in mechanical stress-induced cardiac hypertrophy. Treatment of neonatal rat cardiomyocytes with the C3 exoenzyme, which abrogates Rho functions, suppressed stretch-induced activation of extracellular signal-regulated protein kinases (ERKs). Overexpression of the Rho GDP dissociation inhibitor (Rho-GDI), dominant-negative mutants of RhoA (DNRhoA), or DNRac1 significantly inhibited stretch-induced activation of transfected ERK2. Overexpression of constitutively active mutants of RhoA slightly activated ERK2 in cardiac myocytes. Overexpression of C-terminal Src kinase, which inhibits functions of the Src family of tyrosine kinases, or overexpression of DNRas had no effect on stretch-induced activation of transfected ERK2. The promoter activity of skeletal alpha-actin and c-fos genes was increased by stretch, and these increases were completely inhibited by either cotransfection of Rho-GDI or pretreatment with C3 exoenzyme. Mechanical stretch increased phenylalanine incorporation into cardiac myocytes by approximately 1.5-fold compared with control, and this increase was also significantly suppressed by pretreatment with C3 exoenzyme. Overexpression of Rho-GDI or DNRhoA did not affect angiotensin II-induced activation of ERK. ERKs were activated by culture media conditioned by stretch of cardiomyocytes without any treatment, but not of cardiomyocytes with pretreatment by C3 exoenzyme. These results suggest that the Rho family of small G proteins plays critical roles in mechanical stress-induced hypertrophic responses.  (+info)

Arrest of endosome acidification by bafilomycin A1 mimics insulin action on GLUT4 translocation in 3T3-L1 adipocytes. (3/388)

In insulin-sensitive fat and muscle cells, the major glucose transporter GLUT4 is constitutively sequestered in endosomal tubulovesicular membranes, and moves to the cell surface in response to insulin. While sequence information within GLUT4 appears to be responsible for its constitutive intracellular sequestration, the regulatory elements and mechanisms that enable this protein to achieve its unique sorting pattern under basal and insulin-stimulated conditions are poorly understood. We show here that arrest of endosome acidification in insulin-sensitive 3T3-L1 adipocytes by bafilomycin A1, a specific inhibitor of the vacuolar proton pump, results in the rapid and dose-dependent translocation of GLUT4 from the cell interior to the membrane surface; the effects of maximally stimulatory concentrations of bafilomycin A1 (400-800 nM) were equivalent to 50-65% of the effects of acute insulin treatment. Like insulin, bafilomycin A1 induced the redistribution of GLUT1 and Rab4, but not that of other proteins whose membrane localization has been shown to be insulin-insensitive. Studies to address the mechanism of this effect demonstrated that neither autophosphorylation nor internalization of the insulin receptor was altered by bafilomycin A1 treatment. Bafilomycin-induced GLUT4 translocation was not blocked by cell pretreatment with wortmannin. Taken together, these data indicate that arrest of endosome acidification mimics insulin action on GLUT4 and GLUT1 translocation by a mechanism distal to insulin receptor and phosphatidylinositol 3-kinase activation, and suggest an important role for endosomal pH in the membrane dynamics of the glucose transporters.  (+info)

Molecular dissection of guanine nucleotide dissociation inhibitor function in vivo. Rab-independent binding to membranes and role of Rab recycling factors. (4/388)

Guanine nucleotide dissociation inhibitor (GDI) is an essential protein required for the recycling of Rab GTPases mediating the targeting and fusion of vesicles in the exocytic and endocytic pathways. Using site-directed mutagenesis of yeast GDI1, we demonstrate that amino acid residues required for Rab recognition in vitro are critical for function in vivo in Saccharomyces cerevisiae. Analysis of the effects of Rab-binding mutants on function in vivo reveals that only a small pool of recycling Rab protein is essential for growth, and that the rates of recycling of distinct Rabs are differentially sensitive to GDI. Furthermore, we find that membrane association of Gdi1p is Rab-independent. Mutant Gdi1 proteins unable to bind Rabs were able to associate with cellular membranes as efficiently as wild-type Gdi1p, yet caused a striking loss of the endogenous cytosolic Gdi1p-Rab pools leading to dominant inhibition of growth when expressed at levels of the normal, endogenous pool. These results demonstrate a potential role for a new recycling factor in the retrieval of Rab-GDP from membranes, and illustrate the importance of multiple effectors in regulating GDI function in Rab delivery and retrieval from membranes.  (+info)

Cleavage and nuclear translocation of the caspase 3 substrate Rho GDP-dissociation inhibitor, D4-GDI, during apoptosis. (5/388)

While investigating endonucleases potentially involved in apoptosis, an antisera was raised to bovine deoxyribonuclease II, but it recognized a smaller protein of 26 kDa protein in a variety of cell lines. The 26 kDa protein underwent proteolytic cleavage to 22 kDa concomitantly with DNA digestion in cells induced to undergo apoptosis. Sequencing of the 26 kDa protein identified it as the Rho GDP-dissociation inhibitor D4-GDI. Zinc, okadaic acid, calyculin A, cantharidin, and the caspase inhibitor z-VAD-fmk, all prevented the cleavage of D4-GDI, DNA digestion, and apoptosis. The 26 kDa protein resided in the cytoplasm of undamaged cells, whereas following cleavage, the 22 kDa form translocated to the nucleus. Human D4-GDI, and D4-GDI mutated at the caspase 1 or caspase 3 sites, were expressed in Chinese hamster ovary cells which show no detectable endogenous D4-GDI. Mutation at the caspase 3 site prevented D4-GDI cleavage but did not inhibit apoptosis induced by staurosporine. The cleavage of D4-GDI could lead to activation of Jun N-terminal kinase which has been implicated as an upstream regulator of apoptosis in some systems. However, the results show that the cleavage of D4-GDI and translocation to the nucleus do not impact on the demise of the cell.  (+info)

Phosphoinositide-dependent activation of Rho A involves partial opening of the RhoA/Rho-GDI complex. (6/388)

Rho GTPases have two interconvertible forms and two cellular localizations. In their GTP-bound conformation, they bind to the cell membrane and are activated. In the inactive GDP-bound conformation, they associate with a cytosolic protein called GDP dissociation inhibitor (GDI). We previously reported that the RhoA component of the RhoA/Rho-GDI complex was not accessible to the Clostridium botulinum C3 ADP-ribosyl transferase, unless the complex had been incubated with phosphoinositides. We show here that PtdIns, PtdIns4P, PtdIns3,4P2, PtdIns4,5P2 and PtdInsP3 enhance not only the C3-dependent ADP-ribosylation, but also the GDP/GTP exchange in the RhoA component of the prenylated RhoA/Rho-GDI complex. In contrast, in the nonprenylated RhoA/Rho-GDI complex, the levels of ADP-ribosylation and GDP/GTP exchange are of the same order as those measured on free RhoA and are not modified by phosphoinositides. In both cases, phosphoinositides partially opened, but did not fully dissociate the complex. Upon treatment of the prenylated RhoA/Rho-GDI complex with phosphoinositides, a GTP-dependent transfer to neutrophil membranes was evidenced. Using an overlay assay with the prenylated RhoA/Rho-GDI complex pretreated with PtdIns4P and labeled with [alpha32P]GTP, three membrane proteins with molecular masses between 26 and 32 kDa were radiolabeled. We conclude that in the presence of phosphoinositides, the prenylated RhoA/Rho-GDI complex partially opens, which allows RhoA to exchange GDP for GTP. The opened GTP-RhoA/Rho-GDI complex acquires the capacity to target specific membrane proteins.  (+info)

Rab6 is phosphorylated in thrombin-activated platelets by a protein kinase C-dependent mechanism: effects on GTP/GDP binding and cellular distribution. (7/388)

In platelets and other secretory cells, protein kinase C (PKC) plays a role in exocytosis stimulated by physiological extracellular signals, although its linkage to the secretory machinery is poorly understood. We investigated whether Rab6, a GTP-binding protein that fractionates with platelet alpha-granules, may be involved in linking these processes. We found that Rab6 contains two PKC consensus phosphorylation sites that are evolutionarily conserved. In platelets metabolically labelled with [(32)P]P(i), Rab6 phosphorylation was induced by phorbol esters or by thrombin. This phosphorylation was blocked by a specific PKC inhibitor (Ro-31-8220), but not by a p38 mitogen-activated protein kinase inhibitor (PD-169316). Physiological stimulation of platelets caused a PKC-dependent translocation of Rab6 from platelet particulate fractions, nearly doubling the fraction of Rab6 in the cytosol. A human Rab6 isoform (Rab6C) that is preferentially expressed in human platelet RNA was cloned and its phosphorylation by PKC was characterized. Rab6C incorporated up to 2 mol of [(32)P]P(i) per mol of active protein. Rab6C bound GDP and GTP with K(d) values of 113+/-12 and 119+/-27 nM respectively, and hydrolysed GTP at a rate of 100+/-15 micromol of GTP/mol of Rab6C per min. PKC phosphorylation of Rab6C increased the affinity for GTP by 3-fold, although it had lesser effects on GDP (1.6-fold). Phosphorylation did not alter the GTPase activity. In summary, thrombin activation of platelets leads to PKC-dependent phosphorylation of Rab6 and a translocation of Rab6 to the cytosol. We suggest that PKC phosphorylation may be an important mechanism through which Rab functional interactions in vesicle trafficking and secretion can be altered in response to an external stimulus.  (+info)

Neurite extension occurs in the absence of regulated exocytosis in PC12 subclones. (8/388)

We have investigated the process leading to differentiation of PC12 cells. This process is known to include extension of neurites and changes in the expression of subsets of proteins involved in cytoskeletal rearrangements or in neurosecretion. To this aim, we have studied a PC12 clone (trk-PC12) stably transfected with the nerve growth factor receptor TrkA. These cells are able to undergo both spontaneous and neurotrophin-induced morphological differentiation. However, both undifferentiated and nerve growth factor-differentiated trk-PC12 cells appear to be completely defective in the expression of proteins of the secretory apparatus, including proteins of synaptic vesicles and large dense-core granules, neurotransmitter transporters, and neurotransmitter-synthesizing enzymes. These results indicate that neurite extension can occur independently of the presence of the neurosecretory machinery, including the proteins that constitute the fusion machine, suggesting the existence of differential activation pathways for the two processes during neuronal differentiation. These findings have been confirmed in independent clones obtained from PC12-27, a previously characterized PC12 variant clone globally incompetent for regulated secretion. In contrast, the integrity of the Rab cycle appears to be necessary for neurite extension, because antisense oligonucleotides against the neurospecific isoform of Rab-guanosine diphosphate-dissociation inhibitor significantly interfere with process formation.  (+info)

Guanine Nucleotide Dissociation Inhibitors (GDI) are a group of proteins that bind to and inhibit the dissociation of guanine nucleotides from small GTPases, which are important regulatory molecules involved in various cellular processes such as signal transduction, vesicle trafficking, and cytoskeleton organization.

GDI's function is to maintain these small GTPases in their inactive state by keeping them bound to guanine nucleotides, specifically GDP (guanosine diphosphate). By doing so, GDIs help regulate the activity of small GTPases and control their subcellular localization.

GDIs have been identified in various organisms, including bacteria, yeast, and mammals. In humans, there are two major types of GDIs: RhoGDI (also known as D4-GDI) and RacGDI (also known as GDI-α). These GDIs play crucial roles in regulating the activity of Rho family GTPases, which are involved in various cellular functions such as cell motility, membrane trafficking, and gene expression.

Overall, Guanine Nucleotide Dissociation Inhibitors are essential regulators of small GTPases, controlling their activity and localization to ensure proper cellular function.

Rho-specific guanine nucleotide dissociation inhibitors (RhoGDI) are a group of proteins that regulate the function of Rho GTPases, which are important signaling molecules involved in various cellular processes such as actin cytoskeleton regulation, gene expression, and cell cycle progression.

RhoGDIs bind to Rho GTPases in their inactive state, preventing them from interacting with guanine nucleotide exchange factors (GEFs) that would activate them. By doing so, RhoGDIs help regulate the spatial and temporal activation of Rho GTPases, ensuring that they are activated only when and where needed in the cell.

RhoGDI proteins have been identified as potential targets for therapeutic intervention in various diseases, including cancer, inflammation, and neurological disorders. Inhibitors of RhoGDI function have been shown to disrupt Rho GTPase signaling and may have therapeutic benefits in these conditions.

Rho Guanine Nucleotide Dissociation Inhibitor alpha (RhoGDIα) is a protein that regulates the Rho family of small GTPases, which are important signaling molecules involved in various cellular processes such as actin cytoskeleton regulation, cell motility, and gene expression.

RhoGDIα functions by binding to and inhibiting the dissociation of GDP from Rho GTPases, thereby keeping them in an inactive state in the cytoplasm. When a signal is received, RhoGDIα releases the Rho GTPase, allowing it to exchange GDP for GTP and become activated. Once activated, the Rho GTPase can then interact with downstream effectors to carry out its functions.

RhoGDIα has been implicated in various physiological and pathological processes, including cancer, inflammation, and neurological disorders.

Rho Guanine Nucleotide Dissociation Inhibitor gamma (Rho-GDIγ) is a protein that regulates the Rho family of small GTPases, which are important signaling molecules involved in various cellular processes such as cytoskeleton regulation, cell motility, and gene expression.

Rho-GDIγ functions by binding to and sequestering Rho GTPases in their inactive GDP-bound state in the cytoplasm, preventing them from interacting with downstream effectors. This helps regulate the spatial and temporal activation of Rho GTPases, ensuring proper signal transduction and cellular responses.

Rho-GDIγ is also known as Rho-GDI3 or Ly-GDI, and it shares structural and functional similarities with other Rho-GDI proteins (Rho-GDIα and Rho-GDIβ). However, Rho-GDIγ has a unique pattern of tissue expression and may have distinct regulatory functions in certain cell types.

Rho Guanine Nucleotide Dissociation Inhibitor beta (RhoGDIβ) is a protein that regulates the Rho family of small GTPases, which are important signaling molecules involved in various cellular processes such as actin cytoskeleton regulation, cell motility, and gene expression.

RhoGDIβ functions by binding to and inhibiting the dissociation of GDP from Rho GTPases, thereby keeping them in an inactive state in the cytoplasm. When a signal is received, RhoGDIβ releases the Rho GTPase, allowing it to bind to GTP and become activated. Activated Rho GTPases then interact with downstream effectors to regulate various cellular responses.

RhoGDIβ has been found to play a role in several diseases, including cancer, where it can contribute to tumor progression by promoting cell migration and invasion. Therefore, RhoGDIβ is an attractive target for the development of new therapies for cancer and other diseases.

Guanosine diphosphate (GDP) is a nucleotide that consists of a guanine base, a sugar molecule called ribose, and two phosphate groups. It is an ester of pyrophosphoric acid with the hydroxy group of the ribose sugar at the 5' position. GDP plays a crucial role as a secondary messenger in intracellular signaling pathways and also serves as an important intermediate in the synthesis of various biomolecules, such as proteins and polysaccharides.

In cells, GDP is formed from the hydrolysis of guanosine triphosphate (GTP) by enzymes called GTPases, which convert GTP to GDP and release energy that can be used to power various cellular processes. The conversion of GDP back to GTP can be facilitated by nucleotide diphosphate kinases, allowing for the recycling of these nucleotides within the cell.

It is important to note that while guanosine diphosphate has a significant role in biochemical processes, it is not typically associated with medical conditions or diseases directly. However, understanding its function and regulation can provide valuable insights into various physiological and pathophysiological mechanisms.

Guanosine triphosphate (GTP) is a nucleotide that plays a crucial role in various cellular processes, such as protein synthesis, signal transduction, and regulation of enzymatic activities. It serves as an energy currency, similar to adenosine triphosphate (ATP), and undergoes hydrolysis to guanosine diphosphate (GDP) or guanosine monophosphate (GMP) to release energy required for these processes. GTP is also a precursor for the synthesis of other essential molecules, including RNA and certain signaling proteins. Additionally, it acts as a molecular switch in many intracellular signaling pathways by binding and activating specific GTPase proteins.

Rho GTP-binding proteins are a subfamily of the Ras superfamily of small GTPases, which function as molecular switches in various cellular signaling pathways. These proteins play crucial roles in regulating diverse cellular processes such as actin cytoskeleton dynamics, gene expression, cell cycle progression, and cell migration.

Rho GTP-binding proteins cycle between an active GTP-bound state and an inactive GDP-bound state. In the active state, they interact with various downstream effectors to regulate their respective cellular functions. Guanine nucleotide exchange factors (GEFs) activate Rho GTP-binding proteins by promoting the exchange of GDP for GTP, while GTPase-activating proteins (GAPs) inactivate them by enhancing their intrinsic GTP hydrolysis activity.

There are several members of the Rho GTP-binding protein family, including RhoA, RhoB, RhoC, Rac1, Rac2, Rac3, Cdc42, and Rnd proteins, each with distinct functions and downstream effectors. Dysregulation of Rho GTP-binding proteins has been implicated in various human diseases, including cancer, cardiovascular disease, neurological disorders, and inflammatory diseases.

CDC42 is a small GTP-binding protein that belongs to the Rho family of GTPases. It acts as a molecular switch, cycling between an inactive GDP-bound state and an active GTP-bound state, and plays a critical role in regulating various cellular processes, including actin cytoskeleton organization, cell polarity, and membrane trafficking.

When CDC42 is activated by Guanine nucleotide exchange factors (GEFs), it interacts with downstream effectors to modulate the assembly of actin filaments and the formation of membrane protrusions, such as lamellipodia and filopodia. These cellular structures are essential for cell migration, adhesion, and morphogenesis.

CDC42 also plays a role in intracellular signaling pathways that regulate gene expression, cell cycle progression, and apoptosis. Dysregulation of CDC42 has been implicated in various human diseases, including cancer, neurodegenerative disorders, and immune disorders.

In summary, CDC42 is a crucial GTP-binding protein involved in regulating multiple cellular processes, and its dysfunction can contribute to the development of several pathological conditions.

GTP-binding protein alpha subunits, Gi-Go, are a type of heterotrimeric G proteins that play a crucial role in signal transduction pathways associated with many hormones and neurotransmitters. These G proteins are composed of three subunits: alpha, beta, and gamma. The "Gi-Go" specifically refers to the alpha subunit of these G proteins, which can exist in two isoforms, Gi and Go.

When a G protein-coupled receptor (GPCR) is activated by an agonist, it undergoes a conformational change that allows it to act as a guanine nucleotide exchange factor (GEF). The GEF activity of the GPCR promotes the exchange of GDP for GTP on the alpha subunit of the heterotrimeric G protein. Once GTP is bound, the alpha subunit dissociates from the beta-gamma dimer and can then interact with downstream effectors to modulate various cellular responses.

The Gi-Go alpha subunits are inhibitory in nature, meaning that they typically inhibit the activity of adenylyl cyclase, an enzyme responsible for converting ATP to cAMP. This reduction in cAMP levels can have downstream effects on various cellular processes, such as gene transcription, ion channel regulation, and metabolic pathways.

In summary, GTP-binding protein alpha subunits, Gi-Go, are heterotrimeric G proteins that play an essential role in signal transduction pathways by modulating adenylyl cyclase activity upon GPCR activation, ultimately influencing various cellular responses through cAMP regulation.

GTP-binding proteins, also known as G proteins, are a family of molecular switches present in many organisms, including humans. They play a crucial role in signal transduction pathways, particularly those involved in cellular responses to external stimuli such as hormones, neurotransmitters, and sensory signals like light and odorants.

G proteins are composed of three subunits: α, β, and γ. The α-subunit binds GTP (guanosine triphosphate) and acts as the active component of the complex. When a G protein-coupled receptor (GPCR) is activated by an external signal, it triggers a conformational change in the associated G protein, allowing the α-subunit to exchange GDP (guanosine diphosphate) for GTP. This activation leads to dissociation of the G protein complex into the GTP-bound α-subunit and the βγ-subunit pair. Both the α-GTP and βγ subunits can then interact with downstream effectors, such as enzymes or ion channels, to propagate and amplify the signal within the cell.

The intrinsic GTPase activity of the α-subunit eventually hydrolyzes the bound GTP to GDP, which leads to re-association of the α and βγ subunits and termination of the signal. This cycle of activation and inactivation makes G proteins versatile signaling elements that can respond quickly and precisely to changing environmental conditions.

Defects in G protein-mediated signaling pathways have been implicated in various diseases, including cancer, neurological disorders, and cardiovascular diseases. Therefore, understanding the function and regulation of GTP-binding proteins is essential for developing targeted therapeutic strategies.

A Ral Guanine Nucleotide Exchange Factor (RalGEF) is a type of enzyme that activates the small GTPase proteins known as Ral by promoting the exchange of GDP for GTP. This activation plays a crucial role in various cellular processes, including cell growth, differentiation, and migration. RalGEFs are often targeted in cancer and other diseases due to their involvement in these important signaling pathways.

RhoA (Ras Homolog Family Member A) is a small GTPase protein that acts as a molecular switch, cycling between an inactive GDP-bound state and an active GTP-bound state. It plays a crucial role in regulating various cellular processes such as actin cytoskeleton organization, gene expression, cell cycle progression, and cell migration.

RhoA GTP-binding protein becomes activated when it binds to GTP, and this activation leads to the recruitment of downstream effectors that mediate its functions. The activity of RhoA is tightly regulated by several proteins, including guanine nucleotide exchange factors (GEFs) that promote the exchange of GDP for GTP, GTPase-activating proteins (GAPs) that stimulate the intrinsic GTPase activity of RhoA to hydrolyze GTP to GDP and return it to an inactive state, and guanine nucleotide dissociation inhibitors (GDIs) that sequester RhoA in the cytoplasm and prevent its association with the membrane.

Mutations or dysregulation of RhoA GTP-binding protein have been implicated in various human diseases, including cancer, neurological disorders, and cardiovascular diseases.

Guanine Nucleotide Exchange Factors (GEFs) are a group of regulatory proteins that play a crucial role in the activation of GTPases, which are enzymes that regulate various cellular processes such as signal transduction, cytoskeleton reorganization, and vesicle trafficking.

GEFs function by promoting the exchange of guanosine diphosphate (GDP) for guanosine triphosphate (GTP) on GTPases. GTP is the active form of the GTPase, and its binding to the GTPase leads to a conformational change that activates the enzyme's function.

In the absence of GEFs, GTPases remain in their inactive GDP-bound state, and cellular signaling pathways are not activated. Therefore, GEFs play a critical role in regulating the activity of GTPases and ensuring proper signal transduction in cells.

There are many different GEFs that are specific to various GTPase families, including Ras, Rho, and Arf families. Dysregulation of GEFs has been implicated in various diseases, including cancer and neurological disorders.

Rap GTP-binding proteins, also known as Ras-associated binding (Rab) proteins, are a large family of small GTPases that play crucial roles in regulating intracellular vesicle trafficking and membrane transport. These proteins function as molecular switches that cycle between an active GTP-bound state and an inactive GDP-bound state. In the active state, Rab proteins interact with various effector molecules to mediate specific steps in vesicle budding, transport, tethering, and fusion.

Rab proteins are involved in several cellular processes, including exocytosis, endocytosis, phagocytosis, autophagy, and Golgi apparatus function. Each Rab protein has a specific subcellular localization and is responsible for regulating distinct steps in membrane trafficking pathways. Dysregulation of Rab GTPases has been implicated in various human diseases, including cancer, neurodegenerative disorders, and infectious diseases.

In summary, Rap GTP-binding proteins are a family of small GTPases that regulate intracellular vesicle trafficking and membrane transport by functioning as molecular switches in specific steps of these processes.

Ral GTP-binding proteins are a subfamily of the Ras superfamily of small GTPases, which are molecular switches that regulate various cellular processes, including signal transduction, membrane trafficking, and cytoskeleton dynamics. Ral proteins exist in two isoforms, RalA and RalB, which bind to and hydrolyze GTP (guanosine triphosphate) and GDP (guanosine diphosphate).

Ral GTP-binding proteins are activated by guanine nucleotide exchange factors (GEFs), which promote the exchange of GDP for GTP, thereby converting Ral proteins into their active state. Once activated, Ral proteins interact with various downstream effectors to regulate diverse cellular functions, such as cell growth, differentiation, survival, and motility.

Ral GTP-binding proteins have been implicated in several human diseases, including cancer, where they contribute to tumor progression and metastasis by promoting cell invasion, migration, and angiogenesis. Therefore, Ral GTP-binding proteins are considered promising targets for the development of novel anti-cancer therapies.

Guanine nucleotides are molecules that play a crucial role in intracellular signaling, cellular regulation, and various biological processes within cells. They consist of a guanine base, a sugar (ribose or deoxyribose), and one or more phosphate groups. The most common guanine nucleotides are GDP (guanosine diphosphate) and GTP (guanosine triphosphate).

GTP is hydrolyzed to GDP and inorganic phosphate by certain enzymes called GTPases, releasing energy that drives various cellular functions such as protein synthesis, signal transduction, vesicle transport, and cell division. On the other hand, GDP can be rephosphorylated back to GTP by nucleotide diphosphate kinases, allowing for the recycling of these molecules within the cell.

In addition to their role in signaling and regulation, guanine nucleotides also serve as building blocks for RNA (ribonucleic acid) synthesis during transcription, where they pair with cytosine nucleotides via hydrogen bonds to form base pairs in the resulting RNA molecule.

Protein binding, in the context of medical and biological sciences, refers to the interaction between a protein and another molecule (known as the ligand) that results in a stable complex. This process is often reversible and can be influenced by various factors such as pH, temperature, and concentration of the involved molecules.

In clinical chemistry, protein binding is particularly important when it comes to drugs, as many of them bind to proteins (especially albumin) in the bloodstream. The degree of protein binding can affect a drug's distribution, metabolism, and excretion, which in turn influence its therapeutic effectiveness and potential side effects.

Protein-bound drugs may be less available for interaction with their target tissues, as only the unbound or "free" fraction of the drug is active. Therefore, understanding protein binding can help optimize dosing regimens and minimize adverse reactions.

GTP (Guanosine Triphosphate) Phosphohydrolases are a group of enzymes that catalyze the hydrolysis of GTP to GDP (Guanosine Diphosphate) and inorganic phosphate. This reaction plays a crucial role in regulating various cellular processes, including signal transduction pathways, protein synthesis, and vesicle trafficking.

The human genome encodes several different types of GTP Phosphohydrolases, such as GTPase-activating proteins (GAPs), GTPase effectors, and G protein-coupled receptors (GPCRs). These enzymes share a common mechanism of action, in which they utilize the energy released from GTP hydrolysis to drive conformational changes that enable them to interact with downstream effector molecules and modulate their activity.

Dysregulation of GTP Phosphohydrolases has been implicated in various human diseases, including cancer, neurodegenerative disorders, and infectious diseases. Therefore, understanding the structure, function, and regulation of these enzymes is essential for developing novel therapeutic strategies to target these conditions.

An amino acid sequence is the specific order of amino acids in a protein or peptide molecule, formed by the linking of the amino group (-NH2) of one amino acid to the carboxyl group (-COOH) of another amino acid through a peptide bond. The sequence is determined by the genetic code and is unique to each type of protein or peptide. It plays a crucial role in determining the three-dimensional structure and function of proteins.

Molecular sequence data refers to the specific arrangement of molecules, most commonly nucleotides in DNA or RNA, or amino acids in proteins, that make up a biological macromolecule. This data is generated through laboratory techniques such as sequencing, and provides information about the exact order of the constituent molecules. This data is crucial in various fields of biology, including genetics, evolution, and molecular biology, allowing for comparisons between different organisms, identification of genetic variations, and studies of gene function and regulation.

Guanine is not a medical term per se, but it is a biological molecule that plays a crucial role in the body. Guanine is one of the four nucleobases found in the nucleic acids DNA and RNA, along with adenine, cytosine, and thymine (in DNA) or uracil (in RNA). Specifically, guanine pairs with cytosine via hydrogen bonds to form a base pair.

Guanine is a purine derivative, which means it has a double-ring structure. It is formed through the synthesis of simpler molecules in the body and is an essential component of genetic material. Guanine's chemical formula is C5H5N5O.

While guanine itself is not a medical term, abnormalities or mutations in genes that contain guanine nucleotides can lead to various medical conditions, including genetic disorders and cancer.

Rho Guanine Nucleotide Exchange Factors (Rho-GEFs) are a group of proteins that play a crucial role in the regulation of intracellular signaling pathways. They function as molecular switches that activate Rho GTPases, which are important regulators of various cellular processes such as cytoskeleton reorganization, gene expression, cell cycle progression, and cell migration.

Rho-GEFs catalyze the exchange of guanosine diphosphate (GDP) for guanosine triphosphate (GTP) on Rho GTPases, leading to their activation. This process is tightly regulated and occurs in response to various extracellular signals, such as hormones, growth factors, and integrin-mediated adhesion. Once activated, Rho GTPases interact with downstream effectors to modulate cell behavior.

There are several families of Rho-GEFs, including the Dbl family, the Vav family, and the Trio family, among others. Each family has distinct structural features and regulatory mechanisms that allow for specificity in Rho GTPase activation and downstream signaling. Dysregulation of Rho-GEFs and Rho GTPases has been implicated in various human diseases, including cancer, neurological disorders, and cardiovascular disease.

Rab3 GTP-binding proteins are a subfamily of the Rab family of small GTPases, which are involved in regulating intracellular vesicle trafficking. These proteins play a crucial role in the regulation of neurotransmitter release at synapses in neurons. They are responsible for mediating the docking and fusion of synaptic vesicles with the presynaptic membrane during exocytosis. Rab3 GTP-binding proteins exist in four isoforms (Rab3A, Rab3B, Rab3C, and Rab3D) that share a high degree of sequence similarity. They cycle between an active GTP-bound state and an inactive GDP-bound state, and their activity is regulated by various accessory proteins, including GTP exchange factors (GEFs) and GTPase-activating proteins (GAPs).

Nucleotides are the basic structural units of nucleic acids, such as DNA and RNA. They consist of a nitrogenous base (adenine, guanine, cytosine, thymine or uracil), a pentose sugar (ribose in RNA and deoxyribose in DNA) and one to three phosphate groups. Nucleotides are linked together by phosphodiester bonds between the sugar of one nucleotide and the phosphate group of another, forming long chains known as polynucleotides. The sequence of these nucleotides determines the genetic information carried in DNA and RNA, which is essential for the functioning, reproduction and survival of all living organisms.

Rab GTP-binding proteins, also known as Rab GTPases or simply Rabs, are a large family of small GTP-binding proteins that play a crucial role in regulating intracellular vesicle trafficking. They function as molecular switches that cycle between an active GTP-bound state and an inactive GDP-bound state.

In the active state, Rab proteins interact with various effector molecules to mediate specific membrane trafficking events such as vesicle budding, transport, tethering, and fusion. Each Rab protein is thought to have a unique function and localize to specific intracellular compartments or membranes, where they regulate the transport of vesicles and organelles within the cell.

Rab proteins are involved in several important cellular processes, including endocytosis, exocytosis, Golgi apparatus function, autophagy, and intracellular signaling. Dysregulation of Rab GTP-binding proteins has been implicated in various human diseases, such as cancer, neurodegenerative disorders, and infectious diseases.

Ras Guanine Nucleotide Exchange Factors (Ras-GEFs) are a group of proteins that play a crucial role in the activation of Ras signaling pathways. Ras is a small GTPase protein that acts as a molecular switch, cycling between an inactive GDP-bound state and an active GTP-bound state.

Ras-GEFs function as catalysts to promote the exchange of GDP for GTP on Ras, thereby promoting its activation. This activation leads to the initiation of various downstream signaling cascades that regulate diverse cellular processes such as proliferation, differentiation, and survival.

Ras-GEFs can be classified into two main families based on their structure and mechanism of action: the Dbl family and the non-Dbl family. The Dbl family members contain a conserved Dbl homology (DH) domain that is responsible for catalyzing the exchange of GDP for GTP on Ras. In contrast, non-Dbl family members use alternative mechanisms to promote Ras activation.

Abnormal regulation of Ras-GEFs has been implicated in various human diseases, including cancer and developmental disorders. Therefore, understanding the function and regulation of Ras-GEFs is essential for developing novel therapeutic strategies to target these diseases.

Rac1 (Ras-related C3 botulinum toxin substrate 1) is a GTP-binding protein, which belongs to the Rho family of small GTPases. These proteins function as molecular switches that regulate various cellular processes such as actin cytoskeleton organization, gene expression, cell proliferation, and differentiation.

Rac1 cycles between an inactive GDP-bound state and an active GTP-bound state. When Rac1 is in its active form (GTP-bound), it interacts with various downstream effectors to modulate the actin cytoskeleton dynamics, cell adhesion, and motility. Activation of Rac1 has been implicated in several cellular responses, including cell migration, membrane ruffling, and filopodia formation.

Rac1 GTP-binding protein plays a crucial role in many physiological processes, such as embryonic development, angiogenesis, and wound healing. However, dysregulation of Rac1 activity has been associated with various pathological conditions, including cancer, inflammation, and neurological disorders.

In the context of medicine and pharmacology, "kinetics" refers to the study of how a drug moves throughout the body, including its absorption, distribution, metabolism, and excretion (often abbreviated as ADME). This field is called "pharmacokinetics."

1. Absorption: This is the process of a drug moving from its site of administration into the bloodstream. Factors such as the route of administration (e.g., oral, intravenous, etc.), formulation, and individual physiological differences can affect absorption.

2. Distribution: Once a drug is in the bloodstream, it gets distributed throughout the body to various tissues and organs. This process is influenced by factors like blood flow, protein binding, and lipid solubility of the drug.

3. Metabolism: Drugs are often chemically modified in the body, typically in the liver, through processes known as metabolism. These changes can lead to the formation of active or inactive metabolites, which may then be further distributed, excreted, or undergo additional metabolic transformations.

4. Excretion: This is the process by which drugs and their metabolites are eliminated from the body, primarily through the kidneys (urine) and the liver (bile).

Understanding the kinetics of a drug is crucial for determining its optimal dosing regimen, potential interactions with other medications or foods, and any necessary adjustments for special populations like pediatric or geriatric patients, or those with impaired renal or hepatic function.

No FAQ available that match "guanine nucleotide dissociation inhibitors"

No images available that match "guanine nucleotide dissociation inhibitors"