2021
DOI: 10.3390/cancers13225861
|View full text |Cite
|
Sign up to set email alerts
|

MAPK Inhibition Requires Active RAC1 Signaling to Effectively Improve Iodide Uptake by Thyroid Follicular Cells

Abstract: The Sodium/Iodide Symporter (NIS) is responsible for the active transport of iodide into thyroid follicular cells. Differentiated thyroid carcinomas (DTCs) usually preserve the functional expression of NIS, allowing the use of radioactive iodine (RAI) as the treatment of choice for metastatic disease. However, a significant proportion of patients with advanced forms of TC become refractory to RAI therapy and no effective therapeutic alternatives are available. Impaired iodide uptake is mainly caused by the def… Show more

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
1
1

Citation Types

0
4
0

Year Published

2022
2022
2024
2024

Publication Types

Select...
3

Relationship

1
2

Authors

Journals

citations
Cited by 3 publications
(4 citation statements)
references
References 42 publications
(62 reference statements)
0
4
0
Order By: Relevance
“…These observations were consistent with AJ integrity being required to sustain NIS permanence at the PM of thyroid cells. To further validate these findings, we repeated these experiments in TPC1-HA-NIS cells co-expressing the halide-sensitive mutant of yellow fluorescent protein (YFP) F46L/H148Q/I152L (HS-YFP), which is quenched by iodide, and that we had previously used successfully to study NIS-mediated iodide transport in these and other thyroid cells [ 10 , 12 ]. As shown in Figure 1 E,F, disruption of AJ integrity through EGTA treatment led to a decrease in the rate of NIS-mediated iodide influx (confirmed by its blocking in the presence of NIS inhibitor ClO 4 − ) comparable to the observed decrease in NIS PM levels ( Figure 1 C,D).…”
Section: Resultsmentioning
confidence: 99%
See 2 more Smart Citations
“…These observations were consistent with AJ integrity being required to sustain NIS permanence at the PM of thyroid cells. To further validate these findings, we repeated these experiments in TPC1-HA-NIS cells co-expressing the halide-sensitive mutant of yellow fluorescent protein (YFP) F46L/H148Q/I152L (HS-YFP), which is quenched by iodide, and that we had previously used successfully to study NIS-mediated iodide transport in these and other thyroid cells [ 10 , 12 ]. As shown in Figure 1 E,F, disruption of AJ integrity through EGTA treatment led to a decrease in the rate of NIS-mediated iodide influx (confirmed by its blocking in the presence of NIS inhibitor ClO 4 − ) comparable to the observed decrease in NIS PM levels ( Figure 1 C,D).…”
Section: Resultsmentioning
confidence: 99%
“…In view of our findings, one could speculate that by compromising cell–cell adhesion, this loss of E-cadherin in advanced TCs could contribute to precluding NIS PM residency. Coupled with a decreased transcriptional expression of the symporter, namely via the hyperactivation of the MAP kinase pathway through NRAS and BRAF mutations [ 12 , 22 ], a reduced PM residency could be sufficient to promote RAI refractoriness in advanced TCs. Initial studies towards RAI resensitization therapy in refractory TC have highlighted that approaches promoting TC epithelial redifferentiation often led to improvements, albeit partial, in RAI uptake by TC cells in vitro and in vivo [ 23 ].…”
Section: Discussionmentioning
confidence: 99%
See 1 more Smart Citation
“…Besides, several factors, including blockade of phosphatidylinositol glycan anchor biosynthesis class U (PIGU) by activating an MAPK signaling pathway, increased leukemia-associated RhoA guanine exchange factor (LARG) owing to PTEN deficiency, induction of NIS proteolysis by valosin-containing protein as a component of endoplasmic reticulum-associated degradation, and overexpression of pituitary tumor-transforming gene 1 (PTTG1)-binding factor (PBF), have been shown to trigger impairment of NIS membrane targeting [ 35 , 36 , 37 , 38 , 39 , 40 , 41 ]. On the other hand, recognition of ADP-ribosylation factor 4 (ARF4) by VAPK motif in the NIS C-terminus, regulation of SRC/RAC1/PAK1/PIP5K/EZRIN pathway, activation of RAC1 via MAPK inhibition, application of transient receptor potential vanilloid type 1 (TRPV1) agonist and others were shown to promote NIS functionality and trafficking towards the cell membrane [ 41 , 42 , 43 , 44 , 45 ]. Recently, an article suggested the role of protospacer adjacent motif (PDZ)—PDZ interaction which means PDZ-domain containing protein SCRIB binds to the carboxy-terminus of NIS to localize at proper basolateral plasma membrane [ 46 ].…”
Section: Discussionmentioning
confidence: 99%