scholarly journals PLEKHA4 Promotes Wnt/β-catenin Signaling-Mediated G1/S Transition and Proliferation in Melanoma

2020 ◽  
Author(s):  
Adnan Shami Shah ◽  
Xiaofu Cao ◽  
Andrew C. White ◽  
Jeremy M. Baskin

ABSTRACTMelanoma patients incur substantial mortality, despite promising recent advances in targeted therapies and immunotherapies. In particular, inhibitors targeting BRAF-mutant melanoma can lead to resistance, and no targeted therapies exist for NRAS-mutant melanoma, motivating the search for additional therapeutic targets and vulnerable pathways. Here, we identify a regulator of Wnt/β-catenin signaling, PLEKHA4, as a factor required for melanoma proliferation and survival. PLEKHA4 knockdown in vitro leads to lower Dishevelled levels, attenuated Wnt/β-catenin signaling, and a block of progression through the G1/S cell cycle transition. In mouse xenograft models, inducible PLEKHA4 knockdown attenuated tumor growth in BRAF- and NRAS-mutant melanomas and synergized with the clinically used inhibitor encorafenib in a BRAF-mutant model. As an E3 ubiquitin ligase regulator with both lipid and protein binding partners, PLEKHA4 presents several opportunities for targeting with small molecules. Our work identifies PLEKHA4 as a promising drug target for melanoma and clarifies a controversial role for Wnt/β-catenin signaling in the control of melanoma proliferation.

2020 ◽  
Author(s):  
Maria Kuzikov ◽  
Elisa Costanzi ◽  
Jeanette Reinshagen ◽  
Francesca Esposito ◽  
Laura Vangeel ◽  
...  

Compound repurposing is an important strategy for the identification of effective treatment options against SARS-CoV-2 infection and COVID-19 disease. In this regard, SARS-CoV-2 main protease (3CL-Pro), also termed M-Pro, is an attractive drug target as it plays a central role in viral replication by processing the viral polyproteins pp1a and pp1ab at multiple distinct cleavage sites. We here report the results of a repurposing program involving 8.7 K compounds containing marketed drugs, clinical and preclinical candidates, and small molecules regarded as safe in humans. We confirmed previously reported inhibitors of 3CL-Pro, and have identified 62 additional compounds with IC50 values below 1 uM and profiled their selectivity towards Chymotrypsin and 3CL-Pro from the MERS virus. A subset of 8 inhibitors showed anti-cytopathic effect in a Vero-E6 cell line and the compounds thioguanosine and MG-132 were analysed for their predicted binding characteristics to SARS-CoV-2 3CL-Pro. The X-ray crystal structure of the complex of myricetin and SARS-Cov-2 3CL-Pro was solved at a resolution of 1.77 Angs., showing that myricetin is covalently bound to the catalytic Cys145 and therefore inhibiting its enzymatic activity.


2017 ◽  
Vol 114 (4) ◽  
pp. 681-686 ◽  
Author(s):  
Glen P. Liszczak ◽  
Zachary Z. Brown ◽  
Samuel H. Kim ◽  
Rob C. Oslund ◽  
Yael David ◽  
...  

Recent advances in the field of programmable DNA-binding proteins have led to the development of facile methods for genomic localization of genetically encodable entities. Despite the extensive utility of these tools, locus-specific delivery of synthetic molecules remains limited by a lack of adequate technologies. Here we combine the flexibility of chemical synthesis with the specificity of a programmable DNA-binding protein by using protein trans-splicing to ligate synthetic elements to a nuclease-deficient Cas9 (dCas9) in vitro and subsequently deliver the dCas9 cargo to live cells. The versatility of this technology is demonstrated by delivering dCas9 fusions that include either the small-molecule bromodomain and extra-terminal family bromodomain inhibitor JQ1 or a peptide-based PRC1 chromodomain ligand, which are capable of recruiting endogenous copies of their cognate binding partners to targeted genomic binding sites. We expect that this technology will allow for the genomic localization of a wide array of small molecules and modified proteinaceous materials.


Science ◽  
2018 ◽  
Vol 362 (6414) ◽  
pp. eaat0572 ◽  
Author(s):  
Quinlan L. Sievers ◽  
Georg Petzold ◽  
Richard D. Bunker ◽  
Aline Renneville ◽  
Mikołaj Słabicki ◽  
...  

The small molecules thalidomide, lenalidomide, and pomalidomide induce the ubiquitination and proteasomal degradation of the transcription factors Ikaros (IKZF1) and Aiolos (IKZF3) by recruiting a Cys2-His2 (C2H2) zinc finger domain to Cereblon (CRBN), the substrate receptor of the CRL4CRBN E3 ubiquitin ligase. We screened the human C2H2 zinc finger proteome for degradation in the presence of thalidomide analogs, identifying 11 zinc finger degrons. Structural and functional characterization of the C2H2 zinc finger degrons demonstrates how diverse zinc finger domains bind the permissive drug-CRBN interface. Computational zinc finger docking and biochemical analysis predict that more than 150 zinc fingers bind the drug-CRBN complex in vitro, and we show that selective zinc finger degradation can be achieved through compound modifications. Our results provide a rationale for therapeutically targeting transcription factors that were previously considered undruggable.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 249-249
Author(s):  
Shannon L Maude ◽  
Sarah K Tasian ◽  
Tiffaney Vincent ◽  
Junior W Hall ◽  
Racquel Collins-Underwood ◽  
...  

Abstract Abstract 249 CRLF2 genomic rearrangements that lead to overexpression have been identified in a subset of children with clinically high-risk B-precursor ALL and are highly associated with activating JAK2 mutations (Harvey et al., Blood 2010; Mullighan et al., PNAS 2009 and Nat Genet 2009). These children frequently respond poorly to current intensive cytotoxic chemotherapy regimens and suffer high rates of relapse and mortality. New therapies for these patients are urgently needed. These leukemias exhibit gene expression profiles similar to those of BCR-ABL1 positive ALL, suggesting aberrant kinase activation. We previously demonstrated aberrant JAK/STAT and PI3K/mTOR signaling in CRLF2-overexpressing ALL cell lines and primary human samples in vitro, and thus hypothesize that inhibition of these hyperactive signaling networks has therapeutic relevance. To further characterize this high-risk subset of ALL, we have established multiple xenograft models of CRLF2-rearranged and JAK2-mutated ALL, providing a robust platform for preclinical testing of signal transduction inhibitors. In this model, primary human ALL samples are intravenously injected into NOD/SCID/γc null (NSG) mice, and engraftment is determined by flow cytometry of peripheral blood for human CD19+/CD45+ blasts. Eighteen of 21 primary cryopreserved specimens provided by the Children's Oncology Group engrafted successfully. In order to biochemically characterize the xenografts, we measured phosphorylation of relevant signal transduction proteins by phosphoflow cytometry. Spleens of mice xenografted with CRLF2-rearranged ALL had uniformly increased surface staining of human CRLF2, a component of the heterodimeric receptor complex for the cytokine, thymic stromal lymphopoeitin (TSLP). In vitro stimulation of the CRLF2-overexpressing ALL xenograft spleens with TSLP induced phosphorylation of STAT5, Akt, S6, and 4EBP1, but not of ERK 1/2. In additon, in vitro JAK inhibition with INCB018424 abrogated TSLP-induced JAK/STAT and PI3K/mTOR signaling. The mTOR inhibitor sirolimus, mTORC1/2 inhibitor PP242, and PI3K/mTOR inhibitor PI-103 potently inhibited phosphorylation of S6 and 4EBP1 in these xenograft specimens. These data suggest that the JAK/STAT and PI3K/mTOR pathways may interact in these CRLF2-overexpressing leukemias. These data led us to hypothesize that inhibition of the PI3K/mTOR or JAK/STAT pathways may represent potential therapeutic targets; therefore, we utilized these very high-risk ALL xenograft models to study novel, targeted therapies. Once xenografts had engrafted with sufficient disease burden to detect >5% peripheral CD19+/CD45+ blasts, mice were randomized to receive the mTOR inhibitor sirolimus, the JAK inhibitor INCB018424, or vehicle for three to four weeks. Disease burden was assessed weekly by flow cytometric determination of CD19+/CD45+ blast count in peripheral blood, and at sacrifice, by spleen CD19+/CD45+ blast count. To assess potential differential efficacy based on CRLF2 overexpression (CRLF2+) and/or JAK2 activating mutations (JAK2mut), we treated mice with each subtype of ALL. Sirolimus induced a significant decrease in peripheral blast count in 7 of 7 primary ALL xenografts tested (2 JAK2mut/CRLF2+ samples, 1 JAK2mut/CRLF2- sample, 2 JAK2wt/CRLF2+ samples, and 2 JAK2wt/CRLF2- samples) and a significant decrease in spleen blast count in 6 of 7 samples tested. The most profound reduction of disease burden was seen in the JAK2mut/CRLF2+ leukemias. In addition, the JAK inhibitor INCB018424 decreased peripheral blast count and spleen blast count in a JAK2mut/CRLF2+ xenograft. We next determined if sirolimus conferred a survival advantage in xenografts of 2 ALL specimens, a robust responder and an intermediate responder to sirolimus by blast count. Sirolimus treatment significantly prolonged survival of both xenografts (63 days vs. 23 days, p=0.0015; 91 days vs. 58 days, p=0.0027). Additional human ALL xenograft studies of INCB018424 and other kinase inhibitors are ongoing. The preclinical in vivo efficacy of sirolimus and INCB018424 suggests that novel, targeted therapies have therapeutic potential in CRLF2-overexpressing ALL. Based in part on these data, both INCB018424 and temsirolimus (a parenteral ester of sirolimus) are currently being investigated in multi-center early phase clinical trials for children with relapsed or refractory leukemias. Disclosures: No relevant conflicts of interest to declare.


2021 ◽  
Vol 11 ◽  
Author(s):  
Yufan Tan ◽  
Xiaoyu Zhong ◽  
Xizhi Wen ◽  
Leyi Yao ◽  
Zhenlong Shao ◽  
...  

Melanoma, the most threatening cancer in the skin, has been considered to be driven by the carcinogenic RAF-MEK1/2-ERK1/2 signaling pathway. This signaling pathway is usually mainly dysregulated by mutations in BRAF or RAS in skin melanomas. Although inhibitors targeting mutant BRAF, such as vemurafenib, have improved the clinical outcome of melanoma patients with BRAF mutations, the efficiency of vemurafenib is limited in many patients. Here, we show that blood bilirubin in patients with BRAF-mutant melanoma treated with vemurafenib is negatively correlated with clinical outcomes. In vitro and animal experiments show that bilirubin can abrogate vemurafenib-induced growth suppression of BRAF-mutant melanoma cells. Moreover, bilirubin can remarkably rescue vemurafenib-induced apoptosis. Mechanically, the activation of ERK-MNK1 axis is required for bilirubin-induced reversal effects post vemurafenib treatment. Our findings not only demonstrate that bilirubin is an unfavorable for patients with BRAF-mutant melanoma who received vemurafenib treatment, but also uncover the underlying mechanism by which bilirubin restrains the anticancer effect of vemurafenib on BRAF-mutant melanoma cells.


Science ◽  
2021 ◽  
Vol 372 (6547) ◽  
pp. 1169-1175
Author(s):  
Konstantin Shatalin ◽  
Ashok Nuthanakanti ◽  
Abhishek Kaushik ◽  
Dmitry Shishov ◽  
Alla Peselis ◽  
...  

Emergent resistance to all clinical antibiotics calls for the next generation of therapeutics. Here we report an effective antimicrobial strategy targeting the bacterial hydrogen sulfide (H2S)–mediated defense system. We identified cystathionine γ-lyase (CSE) as the primary generator of H2S in two major human pathogens, Staphylococcus aureus and Pseudomonas aeruginosa, and discovered small molecules that inhibit bacterial CSE. These inhibitors potentiate bactericidal antibiotics against both pathogens in vitro and in mouse models of infection. CSE inhibitors also suppress bacterial tolerance, disrupting biofilm formation and substantially reducing the number of persister bacteria that survive antibiotic treatment. Our results establish bacterial H2S as a multifunctional defense factor and CSE as a drug target for versatile antibiotic enhancers.


Science ◽  
2020 ◽  
Vol 368 (6497) ◽  
pp. 1386-1392 ◽  
Author(s):  
Isaac A. Klein ◽  
Ann Boija ◽  
Lena K. Afeyan ◽  
Susana Wilson Hawken ◽  
Mengyang Fan ◽  
...  

The nucleus contains diverse phase-separated condensates that compartmentalize and concentrate biomolecules with distinct physicochemical properties. Here, we investigated whether condensates concentrate small-molecule cancer therapeutics such that their pharmacodynamic properties are altered. We found that antineoplastic drugs become concentrated in specific protein condensates in vitro and that this occurs through physicochemical properties independent of the drug target. This behavior was also observed in tumor cells, where drug partitioning influenced drug activity. Altering the properties of the condensate was found to affect the concentration and activity of drugs. These results suggest that selective partitioning and concentration of small molecules within condensates contributes to drug pharmacodynamics and that further understanding of this phenomenon may facilitate advances in disease therapy.


2022 ◽  
Author(s):  
Henriette Andresen ◽  
Cristina Pérez-Ternero ◽  
Jerid Robinson ◽  
Deborah M Dickey ◽  
Adrian J Hobbs ◽  
...  

Natriuretic peptide receptor (NPR)-A (also known as NPR-A, NPR1 or guanylyl cyclase-A, GC-A) is an attractive but challenging target to activate with small molecules. GC-A is activated by endogenous atrial natriuretic peptide (ANP) and B-type natriuretic peptide (BNP), and this activation leads to the production of cyclic guanosine monophosphate (cGMP). This system plays an important role in the regulation of cardiovascular and renal homeostasis. However, utilization of this receptor as a drug target has so far been limited to peptides, even though small molecule modulators allow oral administration and longer half-life. We have identified small molecular allosteric enhancers of GC-A, which strengthened ANP or BNP activation in various in vitro and ex vivo systems. These compounds do not mediate their actions through previously described allosteric binding sites or via known mechanisms of action. In addition, their selectivity and activity are dependent on only one amino acid in GC-A. Our findings show that there is a novel allosteric binding site on GC-A, which can be targeted by small molecules that increase the signaling effects of ANP and BNP.


2021 ◽  
Author(s):  
Rossella Loria ◽  
Valentina Laquintana ◽  
Stefano Scalera ◽  
Rocco Fraioli ◽  
Valentina Caprara ◽  
...  

Abstract Background: Despite the promise of dual BRAF/MEK inhibition as a therapy for BRAF-mutant (BRAF-mut) melanoma, heterogeneous responses have been observed in patients, thus predictors of benefit from therapy are needed. We have previously identified semaphorin 6A (SEMA6A) as a BRAF-mut-associated protein involved in actin cytoskeleton remodeling. The purpose of the present study is to dissect the role of SEMA6A in the biology of BRAF-mut melanoma, and to explore its predictive potential towards dual BRAF/MEK inhibition.Methods: SEMA6A expression was assessed by immunohistochemistry in melanoma cohort RECI1 (N=112) and its prognostic potential was investigated in BRAF-mut melanoma patients from DFCI and TCGA datasets (N=258). The molecular mechanisms regulated by SEMA6A to sustain tumor aggressiveness and targeted therapy resistance were investigated in vitro by using BRAF-mut and BRAF-wt melanoma cell lines, an inducible SEMA6A silencing cell model and a microenvironment-mimicking fibroblasts-coculturing model. Finally, SEMA6A prediction of benefit from dual BRAF/MEK inhibition was investigated in melanoma cohort RECI2 (N=14).Results: Our results indicate higher protein expression of SEMA6A in BRAF-mut compared with BRAF-wt melanoma patients and show that SEMA6A is a prognostic indicator in BRAF-mut melanoma from TCGA and DFCI patients cohorts. In BRAF-mut melanoma cells, SEMA6A coordinates actin cytoskeleton remodeling by the RhoA-dependent activation of YAP and dual BRAF/MEK inhibition by dabrafenib+trametinib induces SEMA6A/RhoA/YAP axis. In microenvironment-mimicking co-culture condition, fibroblasts confer to melanoma cells a proliferative stimulus and protect them from targeted therapies, whereas SEMA6A depletion rescues the efficacy of dual BRAF/MEK inhibition. Finally, in BRAF-mut melanoma patients treated with dabrafenib+trametinib, high SEMA6A predicts shorter recurrence-free interval. Conclusions: Overall, our results indicate that SEMA6A contributes to microenvironment-coordinated evasion of melanoma cells from dual BRAF/MEK inhibition and it might be a good candidate predictor of short-term benefit from dual BRAF/MEK inhibition.


Sign in / Sign up

Export Citation Format

Share Document