Any feedback?
Please rate this page
(literature.php)
(0/150)

BRENDA support

Literature summary for 2.3.2.13 extracted from

  • Katt, W.P.; Antonyak, M.A.; Cerione, R.A.
    Opening up about tissue transglutaminase when conformation matters more than enzymatic activity (2018), Med One, 3, e180011 .
    View publication on PubMedView publication on EuropePMC

Activating Compound

Activating Compound Comment Organism Structure
GDP GTP/GDP and Ca2+ are able to compete with one another to convert tTG from the closed- to open-state, and vice versa Homo sapiens
GTP GTP/GDP and Ca2+ are able to compete with one another to convert tTG from the closed- to open-state, and vice versa Homo sapiens
additional information Arg580 in wild-type tTG forms several hydrogen bonds with GDP, while Arg478 binds only to the terminal phosphate. Arg476 binds more poorly to the terminal phosphate of GDP, and Ser171 does not form hydrogen bonds with the GDP molecule Homo sapiens
thioredoxin-1 thioredoxin-1 is released by macrophages exposed to inflammatory stimuli in sufficient quantity to reduce the tTG C370-C371 disulfide bond, activating the enzyme Homo sapiens

Application

Application Comment Organism
diagnostics tTG is considered a prognostic marker for Parkinson's disease. tTG antibodies, and crosslinked gliadin, act as diagnostic tools in celiac disease Homo sapiens

Cloned(Commentary)

Cloned (Comment) Organism
recombinant expression of enzyme mutants in HeLa cells and in NIH 3T3 fibroblasts, nearly half of the cells die within 24 h. In contrast, introduction of wild-type tTG into those same cells does not induce cell death. Mutant R580L/C277A still induces cell death. Although ectopic expression of tTG mutant R580A in striatal cells is not sufficient to induce cell death, it does so in HeLa or NIH 3T3 cells Homo sapiens

Protein Variants

Protein Variants Comment Organism
C277A the mutant is unable to bind guanine nucleotides Homo sapiens
C277S the mutation has no effect on cell death Homo sapiens
C277V the mutant is susceptible to digestion by trypsin, and significantly impaired in nucleotide binding Homo sapiens
D306N/N310A the purified tTG mutant adopts a conformation similar to that of wild-type tTG, based on their mutual ability to bind bodipy-GTP-gammaS and to resist proteolysis by trypsin Homo sapiens
D434A the mutant can be transiently expressed in NIH 3T3 cells but not be generated as recombinant protein. The mutant is cytotoxic when expressed in NIH 3T3 cells Homo sapiens
F174A the F174A mutant is deficient in nucleotide binding, and is digested by trypsin in the presence of GTP-gammaS, Phe174 appears to be involved in a pi-stacking interaction Homo sapiens
F174W mutant resists proteolysis and is able to bind nucleotide Homo sapiens
K677A the mutant can be transiently expressed in NIH 3T3 cells but not be generated as recombinant protein, the mutant is unable to bind bodipy-GTP-gammaS, and shows high sensitivity to degradation by trypsin. The mutant is cytotoxic when expressed in NIH 3T3 cells Homo sapiens
additional information identification of tTG mutants which adopt either the open or the closed state, overview Homo sapiens
N681A the mutant can be transiently expressed in NIH 3T3 cells and generated as recombinant protein. The mutant is cytotoxic when expressed in NIH 3T3 cells Homo sapiens
Q163D the mutant shows no loss of nucleotide binding ability when assayed with [alpha-32P] GTP, and exhibits only a moderate loss of binding ability when assayed with [35S]GTP-gammaS Homo sapiens
Q163L the mutant shows no loss of nucleotide binding ability when assayed with [alpha-32P] GTP, and exhibits only a moderate loss of binding ability when assayed with [35S]GTP-gammaS Homo sapiens
Q164L the mutant shows no loss of nucleotide binding ability when assayed with [alpha-32P] GTP, and exhibits only a moderate loss of binding ability when assayed with [35S]GTP-gammaS Homo sapiens
Q169L the mutant shows no loss of nucleotide binding ability when assayed with [alpha-32P] GTP, and exhibits only a moderate loss of binding ability when assayed with [35S]GTP-gammaS Homo sapiens
R476A the mutant binds nucleotide as well as the wild-type enzyme Homo sapiens
R478A the mutant has partially reduced nucleotide binding Homo sapiens
R579A the R579A mutant of tTG is far more susceptible to proteolysis by trypsin or by calpain than the wild-type Homo sapiens
R580A the mutant is GTP-binding deficient Homo sapiens
R580K decreases in nucleotide binding are observed for the R580L and R580K mutants Homo sapiens
R580L decreases in nucleotide binding are observed for the R580L and R580K mutants Homo sapiens
R580L/C277A the tTG mutant is deficient in GTP-binding and protein crosslinking activity, but still induces cell death Homo sapiens
S171A the mutant binds nucleotide as well as the wild-type enzyme Homo sapiens
W254A the mutant can be transiently expressed in NIH 3T3 cells and generated as recombinant protein, the mutant is unable to bind bodipy-GTP-gammaS, and shows high sensitivity to degradation by trypsin. W254A forms a dimer of tTG molecules in the open-state conformation. The mutant is cytotoxic when expressed in NIH 3T3 cells Homo sapiens
Y516C the mutant is less capable of binding guanine nucleotide compared to wild-type Homo sapiens
Y516F the mutant is less capable of binding guanine nucleotide compared to wild-type Homo sapiens

Inhibitors

Inhibitors Comment Organism Structure
KCC009 a specific tTG inhibitor Homo sapiens

Localization

Localization Comment Organism GeneOntology No. Textmining
cell surface
-
Homo sapiens 9986
-
endoplasmic reticulum tTG and alpha-synuclein both localize to the endoplasmic reticulum in disease brain samples Homo sapiens 5783
-
extracellular
-
Homo sapiens
-
-
intracellular
-
Homo sapiens 5622
-

Metals/Ions

Metals/Ions Comment Organism Structure
Ca2+ required, three calcium binding sites are thought to exist on tTG, GTP/GDP and Ca2+ are able to compete with one another to convert tTG from the closed- to open-state, and vice versa Homo sapiens

Natural Substrates/ Products (Substrates)

Natural Substrates Organism Comment (Nat. Sub.) Natural Products Comment (Nat. Pro.) Rev. Reac.
alpha-synuclein glutamine + alkylamine Homo sapiens
-
alpha-synuclein N5-alkylglutamine + NH3
-
?
ApoE glutamine + alkylamine Homo sapiens
-
ApoE N5-alkylglutamine + NH3
-
?
gliadin glutamine + alkylamine Homo sapiens
-
gliadin N5-alkylglutamine + NH3
-
?
protein glutamine + alkylamine Homo sapiens
-
protein N5-alkylglutamine + NH3
-
?

Organism

Organism UniProt Comment Textmining
Homo sapiens P21980
-
-

Source Tissue

Source Tissue Comment Organism Textmining
brain
-
Homo sapiens
-
Caco-2 cell Caco-2 intestinal cancer cells express crosslinking-competent tTG on their surface Homo sapiens
-
central nervous system the central nervous system has a tTG splice variant which is termed tTG-short, or tTG-S. tTG-S adopts a conformation similar to the open-state Homo sapiens
-
intestine
-
Homo sapiens
-
neuroblastoma cell
-
Homo sapiens
-
pancreatic cancer cell
-
Homo sapiens
-
SH-SY5Y cell
-
Homo sapiens
-

Substrates and Products (Substrate)

Substrates Comment Substrates Organism Products Comment (Products) Rev. Reac.
alpha-synuclein glutamine + alkylamine
-
Homo sapiens alpha-synuclein N5-alkylglutamine + NH3
-
?
ApoE glutamine + alkylamine
-
Homo sapiens ApoE N5-alkylglutamine + NH3
-
?
gliadin glutamine + alkylamine
-
Homo sapiens gliadin N5-alkylglutamine + NH3
-
?
protein glutamine + alkylamine
-
Homo sapiens protein N5-alkylglutamine + NH3
-
?

Synonyms

Synonyms Comment Organism
Galphah
-
Homo sapiens
TG2
-
Homo sapiens
tissue transglutaminase
-
Homo sapiens
transglutaminase
-
Homo sapiens
tTG
-
Homo sapiens
type 2 transglutaminase
-
Homo sapiens

General Information

General Information Comment Organism
malfunction specific tTG inhibitors, such as KCC009, can block alpha-synuclein aggregation in SH-SY5Y neuroblastoma cells Homo sapiens
additional information intracellular tTG is predominantly in the closed-state, with a small portion being in the crosslinking-competent open-state, while extracellular tTG would adopt the open-state, and be crosslinking-competent. This view is somewhat complicated by the mildly oxidative conditions in the extracellular space. A triad of cysteine residues (Cys 230, Cys 370, and Cys 371) are able to make one of two disulfide bonds (C370-C230, or C370-C371) in oxidative conditions. Either disulfide bond reduces crosslinking catalytic activity, but oxidized tTG maintains a conformation similar to the open-state. Cp4d is a reversible small molecule which has little effect on tTG conformation, while NC9 is a bulkier, irreversible peptidomimetic compound presumed to stabilize tTG in the open-state. Cp4d treatment has little effect on the sensitivity of the assorted cells to glucose-oxygen deprivation-induced cell death. NC9 causes the tTG wild-type and tTG C277S mutant expressing cells to undergo a greater degree of cell death under the same conditions. The conformation of tTG is responsible for the cell death enhancement. Binding partners of tTG depend upon its conformation, confromation-independent binding modes of tTG, structure-function analysis, detailed overview Homo sapiens
physiological function type 2 transglutaminase, tTG, is an important player in numerous diseases, including celiac disease, neuronal degenerative diseases, and cancer, and its roles in these diseases often depend as much upon its conformation as its catalytic activity. Several roles of tTG in diseases, detailed overview. ApoE is able to protect against Abeta plaque formation by transporting Abeta out of the brain. Crosslinking by tTG inactivates the protein, rendering it unable to clear Abeta. Parkinson's disease is a related disorder, and tTG plays similar, but unique, roles in this disease as well. Where Alzheimer's disease is driven in part by a buildup of Abeta aggregates, Parkinson's disease is caused by the aggregation of alpha-synuclein. In Parkinson's disease, alpha-synuclein is incorrectly processed into beta-pleated fibrils, which in turn aggregate to form cytoplasmic inclusions called Lewy Bodies. tTG catalyzes the crosslinking of alpha-synuclein, both in vitro and in cell models, tTG and alpha-synuclein both localize to the endoplasmic reticulum in disease brain samples. tTG is the major autoantigen in celiac disease. Celiac disease is an auto-immune disorder in which T-cells attack and damage the small intestine. This process is driven by gliadin, a protein in most grains, which precipitates an immune response. Crosslinking of gliadin formed antigenic complexes. And thioredoxin-1 is released by macrophages exposed to inflammatory stimuli in sufficient quantity to reduce the tTG C370-C371 disulfide bond, activating the enzyme. Since inflammatory conditions are present in celiac disease gut, this effect essentially creates a self-stimulating loop in which activated tTG leads to inflammation, which then activates more tTG. tTG has been shown to play roles in cancer cell adhesion, migration, and invasion via its interactions with fibronectin. tTG binds to fibronectin, and crosslinks it to various surfaces, allowing cells to adhere. Matrix metalloproteinase can then break these crosslinks, and in combination with tTG crosslinking this allows for cell motility. Similarly, tTG is thought to play a role in vesicle trafficking by helping to dock extracellular vesicles (microvesicles) generated by aggressive cancer cells to fibroblasts, through its ability to bind and crosslink fibronectin on the vesicle surface. This docking event can then be blocked by inhibiting its crosslinking activity. tTG can promote either cell survival or apoptosis, depending upon the physiological context. As a pro-survival protein, the crosslinking-competent, open-state form of tTG has been shown to crosslink pRB (a pro-apoptotic protein), causing it to oligomerize and thus lose its activity. This is analogous to its role in Alzheimer's disease, crosslinking ApoE. Closed-state tTG is able to sequester c-Cbl, and block ubiquitinylation and subsequent degradation of the EGF receptor, thereby also promoting cell growth and survival. Thus, both open- and closed-state can tTG promote survival depending upon the specific conditions. The same is true of its pro-apoptotic functions. In pancreatic cancer cells treated with the calcium ionophore A23187, tTG adopts the crosslinking-active open-state to facilitate release of the apoptosis-inducing factor from mitochondria, promoting cell death. In contrast, ectopically expressed tTG in SH-SY5Y cells, which presumably exists in the closed-state, is found to promote apoptosis following osmotic shock or staurosporine treatment. Cytotoxicity of the open-state of tTG Homo sapiens