scholarly journals Proteasome Regulation by Reversible Tyrosine Phosphorylation at the Membrane

2020 ◽  
Author(s):  
Lu Chen ◽  
Xin Shu ◽  
Qiong Chen ◽  
Tiantian Wei ◽  
Xiaorong Wang ◽  
...  

AbstractReversible phosphorylation has emerged as an important mechanism for regulating 26S proteasome function in health and disease. Over 100 phospho-tyrosine (pTyr) sites of the human proteasome have been detected, and yet their function and regulation remain poorly understood. Here we show that the 19S subunit Rpt2 is phosphorylated at Tyr439, a strictly conserved residue within the C-terminal HbYX motif of Rpt2 that is essential for 26S proteasome assembly. Unexpectedly, we found that Y439 phosphorylation depends on Rpt2 membrane localization mediated by its N-myristoylation. Multiple receptor tyrosine kinases (RTKs) can trigger Rpt2-Y439 phosphorylation by activating Src, a N-myristoylated tyrosine kinase. Src directly phosphorylates Rpt2-Y439 in vitro and negatively regulates 26S proteasome integrity and activity at cellular membranes, which can be reversed by the membrane-associated isoform of protein tyrosine phosphatase non-receptor type 2 (PTPN2). In H1975 lung cancer cells with activated Src, blocking Rpt2-Y439 phosphorylation by the Y439F mutation conferred partial resistance to the Src inhibitor saracatinib both in vitro and in a mouse xenograft tumor model, and caused significant changes of cellular responses to saracatinib at the proteome level. Our study has defined a novel mechanism involved in the spatial regulation of proteasome function and provided new insights into tyrosine kinase inhibitor-based anti-cancer therapies.

1995 ◽  
Vol 268 (1) ◽  
pp. C154-C161 ◽  
Author(s):  
G. Bischof ◽  
B. Illek ◽  
W. W. Reenstra ◽  
T. E. Machen

We studied a possible role of tyrosine kinases in the regulation of Ca entry into colonic epithelial cells HT-29/B6 using digital image processing of fura 2 fluorescence. Both carbachol and thapsigargin increased Ca entry to a similar extent and Ca influx was reduced by the tyrosine kinase inhibitor genistein (50 microM). Further experiments were performed in solutions containing 95 mM K to depolarize the membrane potential, and the effects of different inhibitors on influx of Ca, Mn, and Ba were compared. Genistein, but not the inactive analogue daidzein nor the protein kinase C inhibitor 1-(5-isoquinolinylsulfonyl)-2- methylpiperazine, decreased entry of all three divalent cations by 47-59%. In high-K solutions, carbachol or thapsigargin both caused intracellular Ca to increase to a plateau of 223 +/- 19 nM. This plateau was reduced by the tyrosine kinase inhibitors genistein (to 95 +/- 8 nM), lavendustin A (to 155 +/- 17 nM), and methyl-2,5-dihydroxycinnamate (to 39 +/- 3 nM). Orthovanadate, a protein tyrosine phosphatase inhibitor, prevented the inhibitory effect of genistein. Ca pumping was unaffected by genistein. Carbachol increased tyrosine phosphorylation (immunoblots with anti-phosphotyrosine antibodies) of 110-, 75-, and 70-kDa proteins, and this phosphorylation was inhibited by genistein. We conclude that carbachol and thapsigargin increase Ca entry, and tyrosine phosphorylation of some key proteins may be important for regulating this pathway.


Blood ◽  
1999 ◽  
Vol 93 (5) ◽  
pp. 1707-1714 ◽  
Author(s):  
Michael H. Tomasson ◽  
Ifor R. Williams ◽  
Robert Hasserjian ◽  
Chirayu Udomsakdi ◽  
Shannon M. McGrath ◽  
...  

Abstract The TEL/PDGFβR fusion protein is expressed as the consequence of a recurring t(5;12) translocation associated with chronic myelomonocytic leukemia (CMML). Unlike other activated protein tyrosine kinases associated with hematopoietic malignancies, TEL/PDGFβR is invariably associated with a myeloid leukemia phenotype in humans. To test the transforming properties of TEL/PDGFβR in vivo, and to analyze the basis for myeloid lineage specificity in humans, we constructed transgenic mice with TEL/PDGFβR expression driven by a lymphoid-specific immunoglobulin enhancer-promoter cassette. These mice developed lymphoblastic lymphomas of both T and B lineage, demonstrating that TEL/PDGFβR is a transforming protein in vivo, and that the transforming ability of this fusion is not inherently restricted to the myeloid lineage. Treatment of TEL/PDGFβR transgenic animals with a protein tyrosine kinase inhibitor with in vitro activity against PDGFβR (CGP57148) resulted in suppression of disease and a prolongation of survival. A therapeutic benefit was apparent both in animals treated before the development of overt clonal disease and in animals transplanted with clonal tumor cells. These results suggest that small-molecule tyrosine kinase inhibitors may be effective treatment for activated tyrosine kinase–mediated malignancies both early in the course of disease and after the development of additional transforming mutations.


Blood ◽  
1997 ◽  
Vol 90 (12) ◽  
pp. 4947-4952 ◽  
Author(s):  
Martin Carroll ◽  
Sayuri Ohno-Jones ◽  
Shu Tamura ◽  
Elisabeth Buchdunger ◽  
Jürg Zimmermann ◽  
...  

Abstract CGP 57148 is a compound of the 2-phenylaminopyrimidine class that selectively inhibits the tyrosine kinase activity of the ABL and the platelet-derived growth factor receptor (PDGFR) protein tyrosine kinases. We previously showed that CGP 57148 selectively kills p210BCR-ABL–expressing cells. To extend these observations, we evaluated the ability of CGP 57148 to inhibit other activated ABL tyrosine kinases, including p185BCR-ABL and TEL-ABL. In cell-based assays of ABL tyrosine phosphorylation, inhibition of ABL kinase activity was observed at concentrations similar to that reported for p210BCR-ABL. Consistent with the in vitro profile of this compound, the growth of cells expressing activated ABL protein tyrosine kinases was inhibited in the absence of exogenous growth factor. Growth inhibition was also observed with a p185BCR-ABL–positive acute lymphocytic leukemia (ALL) cell line generated from a Philadelphia chromosome–positive ALL patient. As CGP 57148 inhibits the PDGFR kinase, we also showed that cells expressing an activated PDGFR tyrosine kinase, TEL-PDGFR, are sensitive to this compound. Thus, this compound may be useful for the treatment of a variety of BCR-ABL–positive leukemias and for treatment of the subset of chronic myelomonocytic leukemia patients with a TEL-PDGFR fusion protein.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 1569-1569
Author(s):  
Tereza Radosova Muchova ◽  
Eva Reznickova ◽  
Zuzana Somikova ◽  
Tomas Gucky ◽  
Miroslav Strnad ◽  
...  

Abstract Class III receptor tyrosine kinases (RTK), which include c-fms, c-kit, FMS-like tyrosine kinase receptor-3(FLT3) and platelet-derived growth factor receptor (PDGFR) α/β are expressed on acute myelogenous leukemia (AML) cells from the majority of patients and stimulate survival and proliferation of leukemic blasts. FLT3 activation cooperates, for instance, with oncogenic mixed lineage leukemia (MLL) fusion proteins in MLL-induced transformation. The most common FLT3 activation mutation, internal tandem duplication (ITD), is the most frequently observed molecular defect in AML, and it is associated with early relapses and poor prognosis. FLT3-ITD leads to constitutive, ligand-independent activation of the kinase; this results in FLT3 autophosphorylation and induction of several downstream signaling cascades including Ras/MAPK kinase (MEK)/extracellular signal-regulated kinase (ERK) and STAT5 pathways. FLT3 as well as other class III RTK have been widely accepted as suitable drug targets. Several potent inhibitors have been developed, and some of them, such as quizartinib or crenolanib, have demonstrated promising clinical outcomes. However, resistance to these inhibitors remains a significant clinical problem; therefore, development of novel inhibitors is needed. Also Src family tyrosine kinases (SFK) have been proven as therapeutic targets in multiple cancers including leukemia. Here, we developed and tested a novel series of compounds which revealed dual inhibitory activities against class III RTK and SFK, and tested them in vitro against FLT3- and PDGFRα-mutated leukemic cells and in vivo against FLT3-ITD-positive AML. First, we tested kinase selectivity of the novel compounds. Kinase-inhibitory properties were screened at single concentration of 10 nM in biochemical phosphorylation assays against 300 kinases. The compounds revealed strong and specific inhibitory activity especially against class III RTK and SFK. Then, we have used multiple cell lines harboring various oncogenic kinases to test in vitro growth inhibition potential of the compounds. The most potent newly synthesized inhibitor, designated 3922, showed EC50 values at low nanomolar concentrations against FLT3-ITD-positive cell line MV4-11 and FIP1L1-PDGFRα-positive EOL-1 cells. We also used primary cells derived from mouse bone marrow bearing inducible fusion oncogene MLL-ENL-ER (MEER) adapted to growth in cell culture (Takacova et al, 2012, Cancer Cell 21:517). Prior to drug testing, the MEER cells were grown in cell culture media to induce cytokine addiction either to stem cell factor (SCF) or to ligand of FLT3 (FLT3L). The efficacy of 3922 was 5 times more potent against FLT3L-addicted MEER cells than against SCF-addicted cells suggesting that FLT3 is the main target of 3922. We then compared the effect of 3922 on inhibition of FLT3 phosphorylation (pFLT3-Tyr589/591) with quizartinib (Zarrinkar et al, 2009, Blood 114:2984). Both compounds showed to be very efficient inhibitors of FLT3 phosphorylation at nanomolar concentrations, however, 3922 inhibitory effect had longer durability after drug withdrawal when compared with quizartinib. Finally, we determined the activity of 3922 in vivo. A single-dose of 10 mg/kg of 3922 or quizartinib was administered to the mice with subcutaneously implanted MV4-11 xenograft. Quantitation of pFLT3 revealed that the RTK was inhibited by 95% even after 2 hours administration of 3922 and this inhibition sustained 24 hours, in contrast to elevated pFLT3 24 hours after quizartinib administration (Zarrinkar et al, 2009; Gunawardane et al, 2013, Mol Cancer Ther 12:438). After 2 hours, the pERK1/2 levels were reduced by both inhibitors, however, returned to phosphorylation levels comparable to vehicle treated control in 24 hours after administration. The inhibitory effect was more pronounced on phosphorylation of STAT5. Inhibitor 3922 reduced the pSTAT5 level by more than 95% after 24 hours, slightly more effectively than quizartinib. Induction of apoptosis was assessed by PARP cleavage. Cleaved PARP was significantly elevated by 3922 even after 2 hours of treatment, with a pattern similar to the PARP cleavage induced by quizartinib (Gunawardane et al, 2013). In conclusion, we have developed a novel highly potent tyrosine kinase inhibitor effective against AML in vitro and in vivo. Acknowledgment: Supported by NV15-28951A from Ministry of Health, Czech Republic. Disclosures No relevant conflicts of interest to declare.


2021 ◽  
Author(s):  
Shalini Saxena ◽  
Kranti Meher ◽  
Madhuri Rotella ◽  
Subhramanyam Vangala ◽  
Satish Chandran ◽  
...  

COVID-19 pandemic has ravaged the world and vaccines have been rapidly developed as preventive measures. But there is no target-based therapy which can be used if infection sets in. Remdesiver and dexamethasone were not designed to combat COVID-19 but are used clinically till better targeted therapies are available. Given this situation target based therapies that intervene in the disease pathway are urgently needed. Since COVID-19 genesis is driven by uncontrolled inflammation and thrombosis and protein kinases are critical in mounting this response, we explored if available tyrosine kinase inhibitors (TKI) can be used as intervention. We profiled four TKI namely Lapatinib, Dasatinib, Pazopanib and Sitravatinib which inhibit tyrosine kinases but are completely distinct in their chemical structures. We demonstrate using in silico and an in vitro 3D-human vascular lung model which profiles anti-inflammatory and anti-thrombogenic properties that all four TKI are active in varying degrees. Our findings that chemically different TKI which share kinase inhibition as the common mechanism of action are active, strongly indicates that it is a tyrosine kinase target-based activity and not off-target arbitrary effect. We propose that TKI, approved for human use and widely available, can be rapidly deployed as specific target-based therapy for COVID-19.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 2862-2862
Author(s):  
Geoffrey A. Bartholomeusz ◽  
Nichalos Donato ◽  
Zeev Estrov ◽  
Waldemar Priebe ◽  
Moshe Talpaz

Abstract Chronic myelogenous leukemia (CML) is a clonal myeloproliferative disorder of hematopoietic stem cells caused by 9:22 reciprocal chromosomal translocation resulting in expression of a highly stable, constitutively active tyrosine kinase, Bcr/Abl. Inhibition of Bcr/Abl with imatinib mesylate, a potent Abl-specific tyrosine kinase inhibitor, is a highly effective therapy for this disease. However, clinical resistance occurs particularly in the later stages of the disease due mainly to the occurrence of point mutations in the Abl kinase domain and continual Bcr/Abl signaling. Alternative and supplemental therapies are still needed and of great clinical interest. Screening a library of small molecular weight compounds for their ability to induce the degradation of critical transcription factors and tyrosine kinases led to the design and synthesis of WP1130, a compound that reduces Jak2 and Bcr/Abl protein stability. Treatment with WP1130 caused the rapid destruction of Bcr/Abl in human CML cell lines (K562, BV173) as well as in clinical CML specimens obtained from patients. This treatment also resulted in dephosphorylation of pSTAT5, pHck and pCrkL, three known targets of Bcr/Abl. Loss of Bcr/Abl protein following WP1130 treatment was similar to that caused by Bcr/Abl silencing (siRNA), and both treatments lead to the induction of apoptosis. Treatment with WP1130 caused rapid degradation of both wild-type and mutant (T315I) Bcr/Abl protein in BaF3 cell expressing these proteins and in a CML cell line expressing wildtype (BV173) and T315I mutant BCR-ABL (BV-173R). Treatment with WP1130 also strongly inhibited colony formation in soft agar of human CML cells from imatinib resistant patients expressing the T315I mutation. The degradation of Bcr/Abl in cells treated with WP1130 was rapid, (1h) and independent of the proteasomal system and HSP90, a chaperone associated with Bcr/Abl stability. Interestingly, WP1130-induced Bcr/Abl degradation was blocked in the presence of vanadate, a tyrosine phosphatase inhibitor. Together, these observations suggest that WP1130-induced Bcr/Abl degradation is mediated by a unique, proteasomal independent pathway in CML cells. Further development of WP1130 is underway with the goal of developing an effective therapy for treating CML by targeting Bcr/Abl protein expressed in early CML progenitors or stem cell populations.


Blood ◽  
1999 ◽  
Vol 93 (5) ◽  
pp. 1707-1714 ◽  
Author(s):  
Michael H. Tomasson ◽  
Ifor R. Williams ◽  
Robert Hasserjian ◽  
Chirayu Udomsakdi ◽  
Shannon M. McGrath ◽  
...  

The TEL/PDGFβR fusion protein is expressed as the consequence of a recurring t(5;12) translocation associated with chronic myelomonocytic leukemia (CMML). Unlike other activated protein tyrosine kinases associated with hematopoietic malignancies, TEL/PDGFβR is invariably associated with a myeloid leukemia phenotype in humans. To test the transforming properties of TEL/PDGFβR in vivo, and to analyze the basis for myeloid lineage specificity in humans, we constructed transgenic mice with TEL/PDGFβR expression driven by a lymphoid-specific immunoglobulin enhancer-promoter cassette. These mice developed lymphoblastic lymphomas of both T and B lineage, demonstrating that TEL/PDGFβR is a transforming protein in vivo, and that the transforming ability of this fusion is not inherently restricted to the myeloid lineage. Treatment of TEL/PDGFβR transgenic animals with a protein tyrosine kinase inhibitor with in vitro activity against PDGFβR (CGP57148) resulted in suppression of disease and a prolongation of survival. A therapeutic benefit was apparent both in animals treated before the development of overt clonal disease and in animals transplanted with clonal tumor cells. These results suggest that small-molecule tyrosine kinase inhibitors may be effective treatment for activated tyrosine kinase–mediated malignancies both early in the course of disease and after the development of additional transforming mutations.


Blood ◽  
1997 ◽  
Vol 90 (12) ◽  
pp. 4947-4952 ◽  
Author(s):  
Martin Carroll ◽  
Sayuri Ohno-Jones ◽  
Shu Tamura ◽  
Elisabeth Buchdunger ◽  
Jürg Zimmermann ◽  
...  

CGP 57148 is a compound of the 2-phenylaminopyrimidine class that selectively inhibits the tyrosine kinase activity of the ABL and the platelet-derived growth factor receptor (PDGFR) protein tyrosine kinases. We previously showed that CGP 57148 selectively kills p210BCR-ABL–expressing cells. To extend these observations, we evaluated the ability of CGP 57148 to inhibit other activated ABL tyrosine kinases, including p185BCR-ABL and TEL-ABL. In cell-based assays of ABL tyrosine phosphorylation, inhibition of ABL kinase activity was observed at concentrations similar to that reported for p210BCR-ABL. Consistent with the in vitro profile of this compound, the growth of cells expressing activated ABL protein tyrosine kinases was inhibited in the absence of exogenous growth factor. Growth inhibition was also observed with a p185BCR-ABL–positive acute lymphocytic leukemia (ALL) cell line generated from a Philadelphia chromosome–positive ALL patient. As CGP 57148 inhibits the PDGFR kinase, we also showed that cells expressing an activated PDGFR tyrosine kinase, TEL-PDGFR, are sensitive to this compound. Thus, this compound may be useful for the treatment of a variety of BCR-ABL–positive leukemias and for treatment of the subset of chronic myelomonocytic leukemia patients with a TEL-PDGFR fusion protein.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 1522-1522
Author(s):  
Shinya Kimura ◽  
Haruna Naito ◽  
Asumi Yokota ◽  
Yuri Kamitsuji ◽  
Eri Kawata ◽  
...  

Abstract Chemical modifications of imatinib mesylate made with the guidance of molecular modeling yielded several promising compounds. Among them, we selected a compound denoted NS-187 (elsewhere described as CNS-9) on the basis of its affinity to Abl, and also to Lyn, which may be involved in imatinib-resistance (Figure). The most striking structural characteristic of NS-187 is its trifluoromethyl (CF3) group at position 3 of the benzamide ring. The presence of the CF3 group strengthened the hydrophobic interactionss of the molecule with the hydrophobic pocket of Abl. Another possible merit of the CF3 group is that it may fix the conformation of the drug by hindering its rotation at the 4-position of the benzamide ring; as a result, a CF3-bearing molecule may be more potent than more flexible compounds such as imatinib. In fact, NS-187 was 25–55 times more potent than imatinib in vitro and and at least 10 times more potent than in vivo. NS-187 also inhibited the phosphorylation and growth of all Bcr-Abl mutants tested except T315I at physiological concentrations. Another special feature of NS-187, in addition to its increased affinity to Abl is its unique spectrum of inhibitory activity against protein kinases. At a concentration of 0.1 μM, NS-187 inhibited only four of 79 tyrosine kinases, that is, Abl, Arg, Fyn, and Lyn. Notably, at 0.1 μM NS-187 did not inhibit PDGFR, Blk, Src or Yes. The IC50 values of NS-187 for Abl, Src and Lyn were 5.8 nM, 1700 nM and 19 nM, respectively, and those of imatinib were 106 nM, >10,000 nM and 352 nM, respectively. These findings indicate that NS-187 acts as a Bcr-Abl/Lyn inhibitor. In this respect, NS-187 may stand out among other novel Abl tyrosine kinase inhibitors, because BMS-354825 inhibits all members of the Src family, while AMN-107 inhibits none of the Src-family kinases. Our proposed docking models of the NS-187/Abl complex support the notion that NS-187 is more specific for Lyn than for Src. The amino acid at position 252 is either Gln or Cys in Src-family proteins. NS-187 inhibited the Gln252-bearing proteins Abl, Fyn and Lyn but had lower activity against the Cys252-bearing Src and Yes. This is probably because Gln, unlike Cys, readily forms hydrogen bonds. The distinguishing characteristic of NS-187, its high affinity for and specific inhibition of Abl and Lyn, may be useful in the treatment of Bcr-Abl-positive leukemia patients. Figure Figure


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1359-1359
Author(s):  
Bianca F. Goemans ◽  
Christian M. Zwaan ◽  
Desiree de Lange ◽  
Jacqueline Cloos ◽  
Dirk Reinhardt ◽  
...  

Abstract Novel treatment strategies to improve the outcome of pediatric AML are required. Around 30% of pediatric AML patients harbor a mutation in the tyrosine kinases FLT3 (±20%) or KIT (±10%). Patients with FLT3 and KIT mutations have a poor prognosis. It was reported that FLT3/ITD length and allelic ratio (AR) influence prognosis. Tyrosine kinase inhibitors (TKI) are novel drugs specifically targeting activated tyrosine kinases. SU11657 is a selective TKI of FLT3 and KIT. SU11657 is comparable to SU11248 (sunitinib, Sutent®), which is FDA approved for the treatment op gastro-intestinal stromal tumors (GIST) and renal cell carcinoma. In a phase I trial of sunitinib in AML, all 4 patients with FLT3 mutations had complete (n=1) or partial morphologic responses (n=3) compared with only 20% (2/10) of patients with WT FLT3. All responses were of short duration. In this study we investigated whether pediatric AML samples were sensitive to SU11657 in vitro, and whether sensitivity to SU11657 was related to mutations in FLT3 and KIT. We studied 77 pediatric AML samples for FLT3/ITD, FLT3 D835 and KIT exon 8 and 17 mutations. In case of a FLT3/ITD mutation the ITD length and AR were determined. All 77 samples were also tested for in vitro sensitivity to SU11657 using the 4 day MTT assay (concentration range 0.0098 – 10μM). Two measures of sensitivity were calculated: 1. The LC50 value (the concentration at which 50% of the cells is killed); 2. The percentage of cells surviving (CS) at 0.625 μM SU11657. The study population consisted of 49 boys and 28 girls. Fifty-five samples were taken at initial diagnosis and 22 at relapse. Median age at diagnosis was 9.0 years and median white blood cell count 78.8×109/L. FAB types were M0 5×, M1 9×, M2 11×, M3 3×, M4 21×, M5 14×, M7 2× and unknown 12×. A FLT3/ITD mutation was detected in 22/77 (29%), a FLT3 D835 mutation in 6/71 (8%) and a KIT exon 17 mutation in 4/55 samples (7%). No KIT exon 8 mutations were detected. There was an approximately 1000 fold difference in LC50 values between the most sensitive and most resistant sample to SU11657. WT FLT3 and KIT samples were relatively resistant to SU11657 (median CS at 0.625 μM SU11657=91%). However, FLT3/ITD positive samples were significantly more sensitive to SU11657 [median CS at 0.625 μM=66% (p<0.0001)], as well as the FLT3 D835 mutated samples [median CS at 0.625 μM=64% (p=0.004)]. There was no relation between the AR or ITD length and sensitivity to SU11657 (Spearmans ρ=−0.11 (p=0.7) and −0.04 (p=1.0), respectively). The 4 KIT mutated samples also were significantly more sensitive to SU11657 than WT FLT3 and KIT samples [median CS at 0.625 μM=70% (p=0.049)]. In conclusion, there was large interpatient variation in in vitro sensitivity to SU11657. FLT3 and KIT mutated pediatric AML samples were more sensitive to SU11657 than samples with WT FLT3 and KIT. There was no relation between FLT3/ITD AR or ITD length and sensitivity to SU11657. Further clinical evaluation of SU11657 or sunitinib combined with chemotherapy, would be of interest since a third of pediatric AML patients potentially would be sensitive.


Sign in / Sign up

Export Citation Format

Share Document