RAD50-interacting protein 1 (RINT1)

The protein contains 792 amino acids for an estimated molecular weight of 90632 Da.

 

Involved in regulation of membrane traffic between the Golgi and the endoplasmic reticulum (ER); the function is proposed to depend on its association in the NRZ complex which is believed to play a role in SNARE assembly at the ER. May play a role in cell cycle checkpoint control (PubMed:11096100). Essential for telomere length control (PubMed:16600870). (updated: Sept. 12, 2018)

Protein identification was indicated in the following studies:

  1. Bryk and co-workers. (2017) Quantitative Analysis of Human Red Blood Cell Proteome. J Proteome Res. 16(8), 2752-2761.
  2. Chu and co-workers. (2018) Quantitative mass spectrometry of human reticulocytes reveal proteome-wide modifications during maturation. Br J Haematol. 180(1), 118-133.

Methods

The following articles were analysed to gather the proteome content of erythrocytes.

The gene or protein list provided in the studies were processed using the ID mapping API of Uniprot in September 2018. The number of proteins identified and mapped without ambiguity in these studies is indicated below.
Only Swiss-Prot entries (reviewed) were considered for protein evidence assignation.

PublicationIdentification 1Uniprot mapping 2Not mapped /
Obsolete
TrEMBLSwiss-Prot
Goodman (2013)2289 (gene list)227853205992269
Lange (2014)123412347281224
Hegedus (2015)2638262202352387
Wilson (2016)165815281702911068
d'Alessandro (2017)18261817201815
Bryk (2017)20902060101081942
Chu (2018)18531804553621387

1 as available in the article and/or in supplementary material
2 uniprot mapping returns all protein isoforms as one entry

The compilation of older studies can be retrieved from the Red Blood Cell Collection database.

The data and differentiation stages presented below come from the proteomic study and analysis performed by our partners of the GReX consortium, more details are available in their published work.

No sequence conservation computed yet.

Interpro domains
Total structural coverage: 0%
Model score: 39

(right-click above to access to more options from the contextual menu)

VariantDescription
dbSNP:rs11556986
dbSNP:rs35971380
dbSNP:rs34310648
ILFS3
ILFS3

The reference OMIM entry for this protein is 610089

Rad50-interacting protein 1; rint1

CLONING

Using a C-terminal fragment of RAD50 (604040) as bait in a yeast 2-hybrid screen of a human B-cell cDNA library, Xiao et al. (2001) cloned RINT1. The deduced 792-amino acid protein has several leucine heptad repeats that form an N-terminal coiled-coil region. Western blot analysis of human cell lines detected an 87-kD RINT1 doublet. Further experiments suggested that the higher molecular mass protein may be translated from an upstream non-AUG start codon. By immunohistochemical analysis, Lin et al. (2007) found that RINT1 was expressed in endoplasmic reticulum (ER), Golgi apparatus, and centrosomes of human HeLa and U2OS cells.

MAPPING

By genomic sequence analysis, Xiao et al. (2001) mapped the RINT1 gene to chromosome 7q22.1.

GENE FUNCTION

Using truncated proteins, Xiao et al. (2001) found that the second leucine heptad repeat of RAD50 and amino acids 257 to 792 of RINT1 were required for RAD50-RINT1 interaction. RINT1 bound RAD50 only during late S and G2/M phases, and human breast cancer cells expressing an N-terminally truncated RINT1 protein displayed defective radiation-induced G2/M checkpoint. Xiao et al. (2001) concluded that RINT1 may play a role in cell cycle control after DNA damage. Kong et al. (2006) found that RBL2 (180203) and RINT1 were essential for telomere length control in human fibroblasts, with loss of either protein leading to longer telomeres. They proposed that RBL2 forms a complex with RAD50 through RINT1 to block telomerase-independent telomere lengthening. Using RNA interference, Lin et al. (2007) found that depletion of RINT1 in HeLa cells led to loss of pericentriolar positioning and dispersal of the Golgi apparatus, concurrent with centrosome amplification in interphase. In synchronized cells, RINT1 deficiency led to multiple abnormalities upon mitotic entry, including aberrant Golgi dynamics during early mitosis and defective reassembly at telophase, formation of multiple spindle poles, and missegregation of chromosomes. Mitotic cells underwent cell death due to overwhelming cellular defects. Lin et al. (2007) concluded that RINT1 is essential to maintain the dynamic integrity of the Golgi apparatus and the centrosome. Docking and fusion of transport vesicles with target membranes requires membrane tethering factors and membrane fusion factors, or SNAREs (soluble N-ethylmaleimide-sensitive factor attachment protein receptors). Arasaki et al. (2013) stated that RINT1 functions as a component of an ER tethering complex that interacts with SNAREs at the ER. This tethering complex also includes ZW10 (603954) and NAG (NBAS; 608025). Using immunoprecipitation and binding studies, Arasaki et al. (2013) found that RINT1 also interacted with the octameric COG complex (see COG1, 606973) at the trans-Golgi network (TGN), and that the COG complex bound SNAREs at the TGN. At the TGN, RINT1 interacted directly with the COG component COG1 and with the SNARE component syntaxin-16 (STX16; 603666). The same N-terminal domain of RINT1 was required for interaction with ZW10 at the ER and with COG1 at the TGN, suggesting a switching mechanism. Knockdown of RINT1 in human cell lines inhibited endosome-to-trans-Golgi vesicle trafficking and caused redistribution of TGN marker proteins to endosomes.

ANIMAL MODEL

Lin et al. (2007) found that homozygous deletion of Rint1 was lethal at embryonic day 5 to 6 in mouse and caused failure of blastocyst outgrowth ex vivo. Approximately ... More on the omim web site

Subscribe to this protein entry history

June 30, 2020: Protein entry updated
Automatic update: OMIM entry 610089 was added.

Feb. 23, 2019: Protein entry updated
Automatic update: model status changed

Oct. 19, 2018: Additional information
Initial protein addition to the database. This entry was referenced in Bryk and co-workers. (2017).