scholarly journals Activated tumor cell integrin αvβ3 cooperates with platelets to promote extravasation and metastasis from the blood stream

2016 ◽  
Vol 140 ◽  
pp. S27-S36 ◽  
Author(s):  
Martin R. Weber ◽  
Masahiko Zuka ◽  
Mihaela Lorger ◽  
Mario Tschan ◽  
Bruce E. Torbett ◽  
...  
2021 ◽  
Vol 5 (Supplement_1) ◽  
pp. A1012-A1013
Author(s):  
Kavitha Godugu ◽  
Hung-yun Lin ◽  
Shaker A Mousa ◽  
Paul J Davis

Abstract Tetraiodothyroaetic acid (tetrac) is a derivative of L-thyroxine with anticancer properties. By multiple molecular mechanisms, tetrac and chemically-modified tetrac induce apoptosis in a variety of human cancer cells in vitro and in xenografts. The anticancer activities of tetrac are initiated at the thyroid hormone analogue receptor on the extracellular domain of plasma membrane integrin αvβ3 (PJ Davis et al., Physiol Rev 101:319-352, 2021). Induction of apoptosis in glioblastoma xenograft with chemically modified tetrac (P-bi-TAT) has yielded 90% in volume of grafts that continues after discontinuation of tetrac. In the present study, we show that human glioblastoma xenograft shrinkage in response to P-bi-TAT is associated with local appearance of phagocytic monocytes and clearance of apoptotic debris (efferocytosis). Primary culture xenograft of glioblastoma cells (GBM 052814, kindly provided by the University of Pittsburgh Medical Center, Department of Neurosurgery) and U87-luc (ATCC, Manassas, VA) xenografts were generated in 5-member groups of nude mice for each tumor cell type and for controls. Five days post-implantation, injection of animals was begun with PBS (control) or P-bi-TAT (10 mg/kg body weight). Injection was continued X21 days and animals were then maintained off-treatment for an additional 21 days. Tumors were harvested, formalin-fixed and slide-mounted, then analyzed by TUNEL assay for apoptosis and by anti-CD68 staining for monocytic macrophage content. Histologic analysis (H&E staining) was also carried out. TUNEL analysis and histopathology of both xenograft models revealed more than 90% apoptotic change with 21-days of P-bi-TAT treatment (P <0.001) and persistence of 40% apoptotic change 3 weeks post-discontinuation of drug (P<0.001 vs. end of treatment change). By H&E histology and CD68 analysis, monocytes accounted for more than 90% of the viable cells after 3 weeks’ drug treatment. Sixty percent of the end-of-treatment monocyte population persisted 3 weeks after discontinuation of P-bi-TAT (P <0.001). Histology revealed negligible cell debris after 3 weeks of drug treatment and at 3 weeks post-discontinuation of P-bi-TAT. Thus, the anticancer/pro-apoptotic action of tetrac-containing P-bi-TAT is associated with efferocytosis that contributes to the frank tumor shrinkage that results from P-bi-TAT treatment of human glioblastoma xenografts. This is the first documentation of efferocytosis regulated from the thyroid hormone analogue receptor on tumor cell integrin αvβ3.


2009 ◽  
Vol 297 (6) ◽  
pp. E1238-E1246 ◽  
Author(s):  
Paul J. Davis ◽  
Faith B. Davis ◽  
Hung-Yun Lin ◽  
Shaker A. Mousa ◽  
Min Zhou ◽  
...  

A thyroid hormone receptor on integrin αvβ3 that mediates cell surface-initiated nongenomic actions of thyroid hormone on tumor cell proliferation and on angiogenesis has been described. Transduction of the hormone signal into these recently recognized proliferative effects is by extracellular-regulated kinases 1/2 (ERK1/2). Other nongenomic actions of the hormone may be transduced by phosphatidylinositol 3-kinase (PI3K) and are initiated in cytoplasm or at the cell surface. PI3K-mediated effects are important to angiogenesis or other recently appreciated cell functions but apparently not to tumor cell division. For those actions of thyroid hormone [l-thyroxine (T4) and 3,3′-5-triiodo-l-thyronine (T3)] that begin at the integrin receptor, tetraiodothyroacetic acid (tetrac) is an inhibitor of and probe for the participation of the receptor in downstream intracellular events. In addition, tetrac has actions initiated at the integrin receptor that are unrelated to inhibition of the effects of T4 and T3 but do involve gene transcription in tumor cells. Discussed here are the implications of translating these nongenomic mechanisms of thyroid hormone analogs into clinical cancer cell biology, tumor-related angiogenesis, and modulation of angiogenesis that is not related to cancer.


Blood ◽  
2014 ◽  
Vol 124 (20) ◽  
pp. 3141-3150 ◽  
Author(s):  
Raphael Leblanc ◽  
Sue-Chin Lee ◽  
Marion David ◽  
Jean-Claude Bordet ◽  
Derek D. Norman ◽  
...  

Key Points ATX stored in α-granules of resting platelets is secreted upon tumor cell-induced aggregation leading to prometastatic LPA production. Nontumoral ATX promotes early bone colonization by breast cancer cells and contributes to the progression of skeletal metastases.


2018 ◽  
Vol 43 (4) ◽  
pp. 215-219 ◽  
Author(s):  
John T. Leith ◽  
Aleck Hercbergs ◽  
Susan Kenney ◽  
Shaker A. Mousa ◽  
Paul J. Davis

2000 ◽  
Vol 275 (7) ◽  
pp. 4796-4802 ◽  
Author(s):  
Tracy A. Shahan ◽  
Abdelilah Fawzi ◽  
Georges Bellon ◽  
Jean-Claude Monboisse ◽  
Nicholas A. Kefalides

1923 ◽  
Vol 38 (4) ◽  
pp. 385-405 ◽  
Author(s):  
Wade H. Brown ◽  
Louise Pearce

Data bearing on the occurrence and distribution of metastases, as recorded in the preceding papers of the series, are analyzed with reference to the parts played by the tumor cell and by conditions which influence cell distribution and cell growth. It is shown that, while the tumor cell is an essential element in the production of metastases, its influence is relatively constant, and that the prime factors responsible for variations in the occurrence and distribution of metastatic growths are conditions that influence cell distribution or that affect the viability of the cell during transport, and conditions that affect the nutrition of the cell wherever it may become lodged. It is further shown that both the presence and the absence of metastases, as well as peculiarities of distribution and of growth, are susceptible of a comparatively simple explanation upon the basis of well recognized principles of pathological action. In brief, it was found that, while the tumor cells might be distributed by either the blood stream or the lymphatics, the mode of distribution and the fate of the cells were determined in accordance with the same general principles that are applicable to the distribution and disposal of foreign cells introduced into the animal body. It was pointed out, however, that in applying these principles to tumor metastasis, it was necessary to recognize the fact that tumor cells bear a definite relation to the tissues of the host and that they are living and agressive agents. Hence, the reaction that they arouse differs in some respects from that produced by inert or unorganized foreign material or by living cells of a foreign species.


2014 ◽  
Vol 28 (S1) ◽  
Author(s):  
John Leith ◽  
Aleck Hercbergs ◽  
Hung‐Yun Lin ◽  
Shaker Mousa ◽  
Heng‐Yuan Tang ◽  
...  

2021 ◽  
Vol 12 ◽  
Author(s):  
Shaker A. Mousa ◽  
Aleck Hercbergs ◽  
Hung-Yun Lin ◽  
Kelly A. Keating ◽  
Paul J. Davis

L-Thyroxine (T4) is the principal ligand of the thyroid hormone analogue receptor on the extracellular domain of integrin αvβ3. The integrin is overexpressed and activated in cancer cells, rapidly dividing endothelial cells, and platelets. The biologic result is that T4 at physiological concentration and without conversion to 3,3’,5-triiodo-L-thyronine (T3) may stimulate cancer cell proliferation and cancer-relevant angiogenesis and platelet coagulation. Pro-thrombotic activity of T4 on platelets is postulated to support cancer-linked blood clotting and to contribute to tumor cell metastasis. We examine some of these findings as they may relate to cancers of the thyroid. Differentiated thyroid cancer cells respond to physiological levels of T4 with increased proliferation. Thus, the possibility exists that in patients with differentiated thyroid carcinomas in whom T4 administration and consequent endogenous thyrotropin suppression have failed to arrest the disease, T4 treatment may be stimulating tumor cell proliferation. In vitro studies have shown that tetraiodothyroacetic acid (tetrac), a derivative of T4, acts via the integrin to block T4 support of thyroid cancer and other solid tumor cells. Actions of T4 and tetrac or chemically modified tetrac modulate gene expression in thyroid cancer cells. T4 induces radioresistance via induction of a conformational change in the integrin in various cancer cells, although not yet established in thyroid cancer cells. The thyroid hormone receptor on integrin αvβ3 mediates a number of actions of T4 on differentiated thyroid cancer cells that support the biology of the cancer. Additional studies are required to determine whether T4 acts on thyroid cancer cells.


Sign in / Sign up

Export Citation Format

Share Document