Replicative Senescence in Good-Risk Acute Myeloid Leukemia.

Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1922-1922
Author(s):  
Mark Zijlmans ◽  
Susan Swiggers ◽  
Maria Rife Soler ◽  
Berna Beverloo

Abstract Immortal cell growth is considered the hallmark of tumor cells. In contrast, normal cells have a limited proliferative capacity of 40–60 cell divisions, also known as the Hayflick limit. The limited proliferative capacity of normal cells relates to gradual telomere shortening as a consequence of the end-replication problem. Upon critical telomere shortening, cells enter a non-replicative but viable state referred to as replicative senescence. These replicative senescent cells stain blue in a beta-Galactosidase assay and activate DNA double-strand break repair pathways at telomeres (e.g. gamma-H2AX foci). In human fibroblast models, escape from senescence results from loss of p53 and Rb function. Escape is associated with reactivation of telomerase. High levels of telomerase, as observed in germ cells and most tumor cells, allow for immortal cell growth. Recently, we demonstrated low levels of telomerase in AML patients with t(8;21) or inv(16) (Swiggers et al, G.C.C. 2006). Interestingly, levels of telomerase in these AML samples were similar to levels of telomerase in normal bone marrow progenitor cells. We hypothesized that AML without re-activated telomerase may still have intact senescence pathways that limit the proliferative capacity of normal cells. This hypothesis was addressed by studying AML patient samples without telomerase re-activation, i.e., t(8;21), t(15;17) or inv(16) (n=10), and a control group of AML with telomerase re-activation (multiple gains/losses of genetic material, n=8). AML samples werelong-time cultured in vitro in the presence of hematopoietic growth factors (range 3–6 weeks),analyzed in vivo following transplantation in NOD-SCID mice andin patients at time of relapse. Cells with all characteristics of replicative senescence, i.e. enlarged, viable, non-proliferating, blue-coloring in beta-Galactosidase assay, critical short telomeres and gamma-H2AX foci at telomeres, were clearly observed in all AML samples with t(8;21), t(15;17) or inv(16). Gradual telomere shortening was observed in these AML cells in vitro upon long-term culture, in vivo after transplantation in NOD-SCID mice and in vivo in patients at relapse compared to time of diagnosis, indicating that these AML cells do not have an adequate telomere maintenance mechanism. We conclude that AML cells with t(8;21), t(15;17) or inv(16) are characterized by intact pathways that induce replicative senescence. Intact pathways that limit proliferative lifespan may be critical to the high cure rates following chemotherapy treatment of patients with good-risk AML.

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Shojiro Katoh ◽  
Atsuki Fujimaru ◽  
Masaru Iwasaki ◽  
Hiroshi Yoshioka ◽  
Rajappa Senthilkumar ◽  
...  

AbstractRegenerative medicine applications require cells that are not inflicted with senescence after in vitro culture for an optimal in vivo outcome. Methods to overcome replicative senescence include genomic modifications which have their own disadvantages. We have evaluated a three-dimensional (3D) thermo-reversible gelation polymer (TGP) matrix environment for its capabilities to reverse cellular senescence. The expression of senescence-associated beta-galactosidase (SA-βgal) by human chondrocytes from osteoarthritis-affected cartilage tissue, grown in a conventional two-dimensional (2D) monolayer culture versus in 3D-TGP were compared. In 2D, the cells de-differentiated into fibroblasts, expressed higher SA-βgal and started degenerating at 25 days. SA-βgal levels decreased when the chondrocytes were transferred from the 2D to the 3D-TGP culture, with cells exhibiting a tissue-like growth until 42–45 days. Other senescence associated markers such as p16INK4a and p21 were also expressed only in 2D cultured cells but not in 3D-TGP tissue engineered cartilage. This is a first-of-its-kind report of a chemically synthesized and reproducible in vitro environment yielding an advantageous reversal of aging of human chondrocytes without any genomic modifications. The method is worth consideration as an optimal method for growing cells for regenerative medicine applications.


2014 ◽  
Author(s):  
Ελένη Γεωργακοπούλου

Ως κυτταρική γήρανση (cellular senescence) ορίζεται η μη αναστρέψιμη παύση του κυτταρικού κύκλου συνεπεία είτε εξάντλησης των τελομερών, είτε κυτταρικού στρες, και θεωρείται μηχανισμός αντίστασης στην καρκινογένεση. Το φαινόμενο της κυτταρικής γήρανσης αποτελεί μια ερευνητική πρόκληση μιας και αποτελεί συνδετικό κρίκο μεταξύ της φυσιολογικής γήρανσης της χρόνιας φλεγμονής και βασικών μονοπατιών της καρκινογένεσης. Ο μέχρι σήμερα πιο αξιόπιστος δείκτης κυτταρικής γήρανσης είναι η ανίχνευση της δραστηριότητας της β- γαλακτοσιδάσης των γηρασμένων κυττάρων ,(senescence-associated-beta-galactosidase SA-β-gal). Η μέθοδος αυτή δεν μπορεί να εφαρμοστεί σε αρχειακό υλικό (ιστούς εγκιβωτισμένους σε παραφίνη) αλλά μόνο σε φρέσκους ιστούς και σε τομές από άμεσα κατεψυγμένους ιστούς (κρυοτομές). Εξαιτίας αυτού του περιορισμού υπάρχει έλλειψη εκτενών κλινικοπαθολογικών μελετών για την κυτταρική γήρανση.ΣΚΟΠΟΣ : Επιχειρήθηκε η αναζήτηση ενός βιολογικού δείκτη κυτταρικής γήρανσης, με εφαρμογή σε αρχειακό υλικό. Επίσης, μελετήθηκε η πρωτότυπη χρησιμοποίησή του ως δείκτη κυτταρικής γήρανσης σε ένα χρόνιο φλεγμονώδες νόσημα που αποτελεί μοντέλο συσχέτισης χρόνιας φλεγμονής και καρκίνου, τον Ομαλό λειχήνα του στόματος.ΥΛΙΚΑ ΚΑΙ ΜΕΘΟΔΟΙ : Αναζητώντας μια μέθοδο ανίχνευσης γηρασμένων κυττάρων εφαρμόσιμη σε αρχειακό υλικό, αξιολογήσαμε την ιστοχημική χρώση Sudan-Black B (SBB), που είναι ειδική για την ανίχνευση της λιποφουσκίνης. Η λιποφουσκίνη είναι ένα συσσωμάτωμα οξειδωμένων πρωτεϊνών, λιπιδίων και μετάλλων, η οποία συσσωρεύεται σε γηρασμένους ιστούς. Αναλύσαμε κυτταρικά συστήματα στα οποία προκλήθηκε κυτταρική γήρανση είτε ύστερα από εξάντληση του πολλαπλασιασμού (replicative senescence) ή από στρεσογόνα ερεθίσματα, προ-καρκινικές αλλοιώσεις σε υπό προϋποθέσεις knock-in ποντίκια που παρουσιάζουν γήρανση, και τέλος σε ανθρώπινες προκαρκινικές αλλοιώσεις που γνωρίζουμε ότι περιέχουν γηρασμένα κύτταρα. Η τεχνική εν συνεχεία εφαρμόστηκε σε δείγματα Ομαλού λειχήνα, Ακανθοκυτταρικού καρκινώματος στόματος , καλοήθεις βλάβες στοματικού βλεννογόνου (ινώματα) και φυσιολογικού στοματικού βλεννογόνου.ΑΠΟΤΕΛΕΣΜΑΤΑ : Στα παραπάνω πειράματα αποδείξαμε την συνταύτιση της λιποφουσκίνης και του SA-β-gal σε in vitro και in vivo γηρασμένα κύττταρα (κατεψυγμένους ιστούς). Tα ευρήματα αυτά συνηγορούν πως η λιποφουσκίνη είναι ένας ικανός υποψήφιος δείκτης κυτταρικής γήρανσης. Επιπρόσθετα, κατεψυγμένοι ιστοί θετικοί για SA-β-gal μονιμοποιήθηκαν σε φορμόλη, εγκλείστηκαν σε παραφίνη και βάφτηκαν με SBB. Οι αντίστοιχες SA-β-gal θετικές περιοχές στον ιστό βάφτηκαν ειδικά για λιποφουσκίνη με SBB, ενώ οι ιστοί που ήταν αρνητικοί για SA-β-gal βρέθηκαν και αρνητικοί για τη λιποφουσκίνη. Τα ευρήματα αυτά ενισχύουν περαιτέρω την ευαισθησία και την ειδικότητα της SBB χρώσης για την ανάδειξη γηρασμένων κυττάρων σε αρχειακό υλικό. Η τελευταία μοναδική ιδιότητα του SBB μπορεί να αξιοποιηθεί για κλινικοπαθολογικές μελέτες στο ευρέως διαθέσιμο αρχειακό υλικό. Επιπρόσθετα, η εφαρμογή της τεχνικής σε τομές παραφίνης από Ομαλό λειχήνα, Ακανθοκυτταρικό καρκίνωμα σε ινώματα και φυσιολογικό ιστό στόματος, ανέδειξε την παρουσία γηρασμένων ινοβλαστών στις τομές των ινωμάτων και του Ομαλού λειχήνα και την απουσία τους στο φυσιολογικό ιστό και το Ακανθοκυτταρικό καρκίνωμα του στόματος. Τα ευρήματα αυτά συνηγορούν υπερ μιας προστατευτικής δράσης αυτών των κυττάρων , πιθανά στα πλαίσια μιας καλοήθους αντίδρασης του στρώματος.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 1841-1841
Author(s):  
Dharminder Chauhan ◽  
Ajita V. Singh ◽  
Arghya Ray ◽  
Teru Hideshima ◽  
Paul G. Richardson ◽  
...  

Abstract Abstract 1841 Introduction: The dimeric Nuclear Factor-kappa B (NF-κB) transcription factor plays a key role during multiple myeloma (MM) cell adhesion-induced cytokine secretion in bone marrow stromal cells, which in turn triggers MM cell growth in a paracrine manner. NF-κB signaling pathway is mediated via canonical (IKK-α/IKK-β/NEMO-P50/65 or NF-κB1) and non-canonical (IKK-α/IKK-α/NIK-p52/RelB or NF-κB2) components. Prior studies have also linked constitutive activation of non-canonical NF-κB pathway to genetic abnormalities/mutation, allowing for an autocrine growth of MM cells. Other recent studies showed that constitutive NF-κB activity in tumor cells from MM patients renders these cells refractory to inhibition by bortezomib; and in fact, that bortezomib induces canonical NF-κB activity. These reports provided the impetus for the development of an agent with ability to modulate canonical and/or non-canonical NF-κB axis, allowing for a more robust and specific inhibition of NF-κB. Recent research and development efforts at Nereus Pharmaceuticals, Inc., have identified a novel small molecule acanthoic acid analog NPI-1342 as a potent NF-κB inhibitor. Here, we examined the effects of NPI-1342 on canonical versus non-canonical NF-κB signaling pathways, as well as its anti-tumor activity against MM cells using both in vitro and in vivo model systems. Methods: We utilized MM.1S, MM.1R, RPMI-8226, U266, KMS12PE, NCI-H929, OCI-MY5, LR5, Dox-40, OPM1, and OPM2 human MM cell lines, as well as purified tumor cells from patients with MM. Cell viability assays were performed using MTT and Trypan blue exclusion assays. Signal transduction pathways were evaluated using immunoblot analysis, ELISA, and enzymology assays. Animal model studies were performed using the SCID-hu model, which recapitulates the human BM milieu in vivo. Results: We first examined the effects of NPI-1342 on lipopolysaccharides (LPS)-induced NF-κB activity. Results showed that NPI-1342 inhibits LPS-stimulated NF-κB activity in vitro, as measured by phosphorylation of IkBa. To determine whether NPI-1342 triggers a differential inhibitory effect on IKKβ versus IKKα, MM.1S MM cells were treated with NPI-1342 for 48 hours, and protein lysates were subjected to kinase activity assays. NPI-1342 blocked IKKα, but not IKKβ or IKKγ phosphorylation. We next assessed whether the inhibitory effect of NPI-1342 on NF-κB activity is associated with cytotoxicity in MM cells. We utilized a panel of MM cell lines: at least five of these have mutations of TRAF3 (MM.1S, MM.1R, DOX40 and U266); one has no known NF-κB mutations (OPM2), and one has amplification of NF-κB1 (OCI-MY5). Treatment of MM cell lines and primary patient (CD138 positive) MM cells for 48 hours significantly decreased their viability (IC50 range 15–20 μM) (P < 0.001; n=3) without affecting the viability of normal peripheral blood mononuclear cells, suggesting selective anti-MM activity and a favorable therapeutic index for NPI-1342. NPI-1342-induced a marked increase in Annexin V+ and PI- apoptotic cell population (P < 0.001, n=3). Mechanistic studies showed that NPI-1342-triggered apoptosis in MM cells is associated with activation of caspase-8, caspase-9, caspase-3, and PARP cleavage. We next examined the in vivo effects of NPI-1342 in human MM xenograft models. For these studies, we utilized the SCID-hu MM model, which recapitulates the human BM milieu in vivo. In this model, MM cells are injected directly into human bone chips implanted subcutaneously in SCID mice, and MM cell growth is assessed by serial measurements of circulating levels of soluble human IL-6R in mouse serum. Treatment of tumor-bearing mice with NPI-1342 (20 mg/kg intraperitoneally, QD1-5 for 2 weeks), but not vehicle alone, significantly inhibits MM tumor growth in these mice (10 mice each group; P = 0.004). The doses of NPI-1342 were well tolerated by the mice, without significant weight loss. Finally, immunostaining of implanted human bone showed robust apoptosis and blockade of NF-κB in mice treated with NPI-1342 versus vehicle alone. Conclusions: We demonstrate the efficacy of a novel small molecule inhibitor of NF-κB NPI-1342 in MM using both in vitro and in vivo models. NPI-1342 blocks NF-κB activity with a preferential inhibitory activity against IKK-α component of NF-κB signaling. Our preclinical studies support evaluation of NPI-1342 as a potential MM therapy. Disclosures: Hideshima: Acetylon: Consultancy. Richardson:Millennium: Membership on an entity's Board of Directors or advisory committees; Celgene: Membership on an entity's Board of Directors or advisory committees; Johnson & Johnson: Membership on an entity's Board of Directors or advisory committees; Novartis: Membership on an entity's Board of Directors or advisory committees; Bristol Myers Squibb: Membership on an entity's Board of Directors or advisory committees. Palladino:Nereus Pharmaceuticals, Inc: Employment, Equity Ownership. Anderson:Celgene: Consultancy; Millennium: Consultancy; Onyx: Consultancy; Merck: Consultancy; Bristol Myers Squibb: Consultancy; Novartis: Consultancy, Membership on an entity's Board of Directors or advisory committees; Acetylon:; Nereus Pharmaceuticals, Inc: Consultancy.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 13086-13086
Author(s):  
S. Ohtani ◽  
K. Ueda ◽  
G. Jayanchandran ◽  
K. Xu ◽  
J. D. Minna ◽  
...  

13086 Background: 101F6 is a candidate tumor suppressor gene on chromosome 3p21.3, a site of allele loss and genomic alterations were frequently found in many human cancers. We previously showed that enforced expression of wt-101F6 by adenoviral virus significantly inhibited tumor cell growth in 3p21.3-deficient NSCLC cells in vitro and in vivo. How 101F6 exerts this effect is largely unknown. Using a computer-aided structural and functional modeling, we recently identified 101F6 as a member of cytochrome b-561 protein family, which is involved in the regeneration of vitamin C. We hypothesized that under normal physiologic conditions, 101F6 protects cells from oxidative damage by regenerating antioxidant vitamin C and that in 101F6-deficient tumor cells, exogenous 101F6 facilitates vitamin C-mediated cytotoxic H2O2 formation. Methods and Results: We examined endogenous 101F6 expression in human NSCLC cell lines and tissue samples. All normal lung bronchial epithelial cells and fibroblasts but few lung cancers expressed 101F6. We investigated the combined effect of 101F6 and vitamin C on the cell growth: a nanoparticle-mediated wt-101F6 gene transfer plus a sub-pharmacologic concentration of vitamin C synergistically inhibited 3p21.3-deficient NSCLC cell growth but did not affect normal cell growth. We also used a human NSCLC H322 orthotopic lung tumor xenograft mouse model to evaluate the therapeutic efficacy of systemic injection of 101F6 nanoparticles and intraperitoneal injection of vitamin C. The growth of lung tumors was synergistically inhibited by the combination treatment (p<0.001). Furthermore, exogenous 101F6 promoted intracellular vitamin C uptake, leading to the vitamin C-mediated accumulation of H2O2 in the tumor cells, and these two agents synergistically killed the cells through caspase-independent apoptosis and autophagy cell death pathways. Conclusions: The synergistic and selective antitumor effect of 101F6 nanoparticles plus vitamin C may offer a useful tool for lung cancer prevention and intervention. This abstract is supported by grants from NCI (SPORE P50CA70907) and DOD (TARGET, DAMD17002–1-0706). No significant financial relationships to disclose.


2006 ◽  
Vol 188 (2) ◽  
pp. 321-331 ◽  
Author(s):  
T Taguchi ◽  
T Takao ◽  
Y Iwasaki ◽  
M Nishiyama ◽  
K Asaba ◽  
...  

Dehydroepiandrosterone (DHEA) is believed to have an anti-tumor effect, as well as anti-inflammatory, antioxidant, and anti-aging effects. To clarify the possible inhibitory action of DHEA on pituitary tumor cells, we tested the effects of DHEA, alone or in combination with the nuclear factor-κB (NF-κB) inhibitor parthenolide (PRT), on AtT20 corticotroph cell growth and function both in vitro and in vivo. We found that, in vitro, DHEA and PRT had potent inhibitory effects on pro-opiomelanocortin and NF-κB-dependent gene expression. They also suppressed the transcription activity of survivin, a representative anti-apoptotic factor, and induced apoptosis in this cell line. Furthermore, using BALB/C nude mice with xenografts of AtT20 cells in vivo, we found that the combined administration of DHEA and PRT significantly attenuated tumor growth and survivin expression. The treatment also decreased the elevated plasma corticosterone levels and ameliorated the malnutrition induced by tumor growth. Altogether, these results suggested that combined treatments of DHEA and PRT potently inhibit the growth and function of corticotroph tumor cells both in vitro and in vivo. This effect may, at least partly, be caused by the suppressive effects of these compounds, such as survivin and other inhibitor of apoptosis proteins, on NF-κB-mediated gene transcription.


eLife ◽  
2021 ◽  
Vol 10 ◽  
Author(s):  
Jessica SY Ho ◽  
Federico Di Tullio ◽  
Megan Schwarz ◽  
Diana Low ◽  
Danny Incarnato ◽  
...  

High spliceosome activity is a dependency for cancer cells, making them more vulnerable to perturbation of the splicing machinery compared to normal cells. To identify splicing factors important for prostate cancer (PCa) fitness, we performed pooled shRNA screens in vitro and in vivo. Our screens identified HNRNPM as a regulator of PCa cell growth. RNA- and eCLIP-sequencing identified HNRNPM binding to transcripts of key homeostatic genes. HNRNPM binding to its targets prevents aberrant exon inclusion and back-splicing events. In both linear and circular mis-spliced transcripts, HNRNPM preferentially binds to GU-rich elements in long flanking proximal introns. Mimicry of HNRNPM dependent linear splicing events using splice-switching-antisense-oligonucleotides (SSOs) was sufficient to inhibit PCa cell growth. This suggests that PCa dependence on HNRNPM is likely a result of mis-splicing of key homeostatic coding and non-coding genes. Our results have further been confirmed in other solid tumors. Taken together, our data reveal a role for HNRNPM in supporting cancer cell fitness. Inhibition of HNRNPM activity is therefore a potential therapeutic strategy in suppressing growth of PCa and other solid tumors.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 639-639
Author(s):  
Masood A. Shammas ◽  
Hemanta Koley ◽  
Pierfrancesco Tassone ◽  
Paola Neri ◽  
Alexei Protopopov ◽  
...  

Abstract Telomerase activity is either low or completely absent in most normal somatic cells; while it is elevated in most cancer cells providing unlimited proliferative potential by preventing telomere shortening. The inhibitors of telomerase, therefore, induce telomere shortening leading to apoptotic cell death in tumor cells while having little or no effect on normal diploid cells. We have evaluated the in vitro and in vivo efficacy of thio-phosphoramidate oligonucleotide specifically targeting the RNA component of telomerase (GRN163L) with demonstrated nuclear uptake by &gt;99% cells without the transfection enhancer. Delivery of GRN163L (1 μM) to MM cells (INA6 and ARP) was specifically associated with complete loss of telomerase activity as early as 6 hrs following exposure and was accompanied by a reduction in myeloma cell growth and survival. Treatment of INA6 cells with GRN163L for three weeks induced 96±4% and 100% cell death at 0.5 and 1 μM concentrations, respectively. ARP cells, which express higher levels of telomerase activity and have longer telomeres, showed 67±4% cell death at 5 weeks with 0.5 μM inhibitor and 82±3% and 100% cell death at 4 and 5 weeks, respectively, with 2 μM GRN163L. The apoptotic cell death was confirmed in 51% INA6 cells at two weeks and in &gt;80% ARP cells at four weeks. Apoptosis was associated with reduction in mean Telomere Fluorescence Intensity (TFI) on interphase chromosomes from 87.1±6.2 in control oligo treated INA6 cells to 36.2±2 (2.4 fold) in GRN163L treated cells. Moreover, GRN163L treatment was also associated with a similar reduction in number of chromosomes with detectable telomeres, indicating development of telomere-free ends. We have confirmed in vivo efficacy of GRN163L in a SCID-hu murine model of multiple myeloma. Following growth of GFP-transduced myeloma cells in the fetal bone chip introduced into the mice, GRN163L was injected on alternate days. In two independent experiments significant reduction in tumor cell growth, as measured by reduction in human myeloma related protein, and better survival than mice injected with control oligo was observed. We have now evaluated efficacy of combination of GRN163L with other novel agents. We have observed synergistic activity with Hsp90 inhibitor 17AAG on myeloma cell death. Addition of 17AAG (0.05 μM) to myeloma cells pre-treated with GRN163L (1 μM) for one week led to complete growth arrest within four days compared to continued growth of cells not pre-treated with GRN-163. These data provide the preclinical rationale for clinical evaluation of GRN163L in myeloma and in combination with Hsp90 inhibitor.


Blood ◽  
2000 ◽  
Vol 95 (10) ◽  
pp. 3133-3138 ◽  
Author(s):  
Jasmine Zain ◽  
Yao-Qi Huang ◽  
XueSheng Feng ◽  
Mary Lynn Nierodzik ◽  
Jian-Jun Li ◽  
...  

Because thrombin-treated tumor cell-induced metastasis increases tumor nodule volume12 greater than nodule number, we studied the effect of thrombin on tumor cell growth in vitro and in vivo (murine B16F10 melanoma, human HCT8 colon carcinoma, DU145 prostate carcinoma). Tumor cell growth was measured after 3 to 7 days in 1% fetal calf serum (FCS) + RPMI 1640. We found that, whereas relatively low concentrations of thrombin, 0.1 to 0.5 U/mL (1-5 nmol/L) enhance tumor cell growth in vitro approximately 2- to 3-fold, higher concentrations, 0.5 to 1 U/mL (5-10 nmol/L) impaired cell growth approximately 2- to 4-fold. Impaired cell growth was associated with cell cycle arrest at G2M and increased pre-GoDNA, as well as apoptosis, measured by tumor cell binding to Annexin V and propidium iodide. Apoptosis was reversed with the general caspase inhibitor, FK-011. The enhancing and inhibiting effects were specific for thrombin (reversed with inactive diisopropyl-fluorophosphate [DFP]-thrombin) and mediated via the protease-activated receptor 1 (PAR-1). PAR-1 activation was demonstrated by (1) use of a cell line, B16F10, devoid of the 3 other thrombin receptors, PAR-3, PAR-4, and GPIb; and (2) greater sensitivity of PAR-1 transfected B16F10 and HCT8 cells to impaired cell growth/apoptosis, 3- and 14-fold, respectively. Thus, thrombin has a bimodal effect on PAR-1 in tumor cells: enhanced growth at low concentration, impaired growth/apoptosis at higher concentration.


Blood ◽  
2000 ◽  
Vol 95 (10) ◽  
pp. 3133-3138 ◽  
Author(s):  
Jasmine Zain ◽  
Yao-Qi Huang ◽  
XueSheng Feng ◽  
Mary Lynn Nierodzik ◽  
Jian-Jun Li ◽  
...  

Abstract Because thrombin-treated tumor cell-induced metastasis increases tumor nodule volume12 greater than nodule number, we studied the effect of thrombin on tumor cell growth in vitro and in vivo (murine B16F10 melanoma, human HCT8 colon carcinoma, DU145 prostate carcinoma). Tumor cell growth was measured after 3 to 7 days in 1% fetal calf serum (FCS) + RPMI 1640. We found that, whereas relatively low concentrations of thrombin, 0.1 to 0.5 U/mL (1-5 nmol/L) enhance tumor cell growth in vitro approximately 2- to 3-fold, higher concentrations, 0.5 to 1 U/mL (5-10 nmol/L) impaired cell growth approximately 2- to 4-fold. Impaired cell growth was associated with cell cycle arrest at G2M and increased pre-GoDNA, as well as apoptosis, measured by tumor cell binding to Annexin V and propidium iodide. Apoptosis was reversed with the general caspase inhibitor, FK-011. The enhancing and inhibiting effects were specific for thrombin (reversed with inactive diisopropyl-fluorophosphate [DFP]-thrombin) and mediated via the protease-activated receptor 1 (PAR-1). PAR-1 activation was demonstrated by (1) use of a cell line, B16F10, devoid of the 3 other thrombin receptors, PAR-3, PAR-4, and GPIb; and (2) greater sensitivity of PAR-1 transfected B16F10 and HCT8 cells to impaired cell growth/apoptosis, 3- and 14-fold, respectively. Thus, thrombin has a bimodal effect on PAR-1 in tumor cells: enhanced growth at low concentration, impaired growth/apoptosis at higher concentration.


Sign in / Sign up

Export Citation Format

Share Document