Morin inhibits STAT3 tyrosine 705 phosphorylation in tumor cells through activation of protein tyrosine phosphatase SHP1

Morin inhibits STAT3 tyrosine 705 phosphorylation in tumor cells through activation of protein tyrosine phosphatase SHP1. Sts-1 we Isorhamnetin 3-O-beta-D-Glucoside miniaturized a phosphatase assay to 1 1,536-well format and conducted a 20,580 compound screen. Among the hits were two classes of structurally related compounds, tetracycline variants and sulfonated azo dyes. These hits had low micromolar to nanomolar IC50 values. Orthogonal screening confirmed the validity of these inhibitors and demonstrated that both act competitively on Sts-1 phosphatase activity. When tested on other PTPs, PTP1B and SHP1, the tetracycline variant, doxycycline, and the sulfonated azo dye, Congo red, are selective inhibitors of Sts-1HP with selectivity indices ranging from 19 to as high as 200. The planar polyaromatic moieties present in both classes of compounds suggested a common binding mode. Mutation of either tryptophan 494 or tyrosine 596, located near the active site of the protein, reduced the of the inhibitors from 3- to 18-fold, indicating that these residues may help promote binding of substrates with aromatic groups. This work provides new insights into substrate selectivity mechanisms and describes two classes of compounds that can serve as probes of function or as a basis for future drug discovery. (6C9). This resistance phenotype is characterized by enhanced survival, rapid clearance of the pathogen and altered inflammatory response. Systemic infections, with close to 50,000 cases per year in the United States alone, have high mortality rates that have not declined in the last 20 years (10C12). mice suggest that therapeutic inactivation of Sts function generates a unique immune response that helps to reduce pathogen burden and prevent the accompanying destructive inflammation. As such, the pharmacological inhibition of Sts activity presents a potential opportunity to generate immuno-stimulatory therapies, to be used in combination with existing standard-of-care antibiotics or antifungals, as a means to treat deadly pathogen infections. Sts-1 and Sts-2, which share 40% sequence identify, are multi-domain proteins composed of an N-terminal ubiquitin association (UBA) domain, a Src-homology 3 (SH3) domain, and a C-terminal histidine phosphatase (HP) domain. The UBA and SH3 domains are believed to mediate protein-protein interactions and may be involved in spatiotemporal regulation of the Sts catalytic function (16C18). The HP domain of these proteins are homologous to the phosphoglycerate mutase (PGM) family, a subgroup of the histidine phosphatase superfamily (5, 19, 20). The HP domain of the Sts proteins, like other members of the PGM family, employs a 2-step catalytic mechanism that involves nucleophilic attack by one of the two conserved histidine residues and formation of a phosphor-histidine intermediate (19, 21C24). While the Sts proteins are known to catalyze the dephosphorylation of phosphotyrosines on protein substrates, they are structurally, functionally and mechanistically distinct from canonical protein tyrosine phosphatases (PTPs) such as PTP1B and SHP1 (5, 20). An established substrate of both Sts-1 and Sts-2, of 1 1 M (20, 26, 27). To identify small molecule inhibitors of Sts-1 for use as functional probes or as a basis for further drug development we miniaturized an established phosphatase assay and conducted a 20,000 compound high throughput screen of the Sts-1 phosphatase domain Sts-1HP. The screen yielded 51 active compounds (IC50 < 10 M) that were inactive in an enzyme-minus counter-screen. Within these hits were several groups of compounds with similar structural scaffolds. Among these, tetracycline analogs and sulfonate-containing azo dyes, were the largest two groups, with more than 10 and 5 variants present in the screening results, respectively. Both compound classes were determined to be competitive inhibitors of Sts-1HP that selectively inhibit this phosphatase, when compared to the PTPs SHP1 and PTP1B. Based on the conserved planar, aromatic structures of these inhibitors we speculated that aromatic residues within the protein were likely involved in critical protein-inhibitor interactions. Consistent with this hypothesis, mutation of.mice suggest that therapeutic inactivation of Sts function generates a unique immune response that helps to reduce pathogen burden and prevent the accompanying destructive inflammation. increases host survival of systemic infection and leads to improved pathogen clearance. The protein tyrosine phosphatase (PTP) activity of these proteins arises from a C-terminal 2-histidine phosphatase (HP) domain. To identify new inhibitors of the HP activity of Mouse monoclonal to HA Tag Sts-1 we miniaturized a phosphatase assay to 1 1,536-well format and conducted a 20,580 compound screen. Among the hits were two classes of structurally related compounds, tetracycline variants and sulfonated azo dyes. These hits had low micromolar to nanomolar IC50 values. Orthogonal screening confirmed the validity of these inhibitors and demonstrated that both act competitively on Sts-1 phosphatase activity. When tested on other PTPs, PTP1B and SHP1, the tetracycline variant, doxycycline, and the sulfonated azo dye, Congo red, are selective inhibitors of Sts-1HP with selectivity indices ranging from 19 to as high as 200. The planar polyaromatic moieties present in both classes of compounds suggested a common binding mode. Mutation of either tryptophan 494 or tyrosine 596, located near the active site of the protein, reduced the of the inhibitors from 3- to 18-fold, indicating that these residues may help promote binding of substrates with aromatic groups. This work provides new insights into substrate selectivity mechanisms and describes two classes of compounds that can serve as probes of function or like a basis for future drug discovery. (6C9). This resistance phenotype is characterized by enhanced survival, rapid clearance of the pathogen and altered inflammatory response. Systemic infections, with close to 50,000 cases per year in the United States alone, have high mortality rates that have not declined in the last 20 years (10C12). mice suggest that therapeutic inactivation of Sts function generates a unique immune response that helps to reduce pathogen burden and prevent the accompanying destructive inflammation. As such, the pharmacological inhibition of Sts activity presents a potential opportunity to generate immuno-stimulatory therapies, to be used in combination with existing standard-of-care antibiotics or antifungals, as a means to treat deadly pathogen infections. Sts-1 and Sts-2, which share 40% sequence identify, are multi-domain proteins composed of an N-terminal ubiquitin association (UBA) domain, a Src-homology 3 (SH3) domain, and a C-terminal histidine phosphatase (HP) domain. The UBA and SH3 domains are believed to mediate protein-protein interactions and may be involved in spatiotemporal regulation of the Sts catalytic function (16C18). The HP domain of those proteins are homologous to the phosphoglycerate mutase (PGM) family, a subgroup of the histidine phosphatase superfamily (5, 19, 20). The HP domain of the Sts proteins, like other members of the PGM family, employs a 2-step catalytic mechanism that involves nucleophilic attack by one of the two conserved histidine residues and formation of a phosphor-histidine intermediate (19, 21C24). While the Sts proteins are known to catalyze the dephosphorylation of phosphotyrosines on protein substrates, they may be structurally, functionally and mechanistically distinct from canonical protein tyrosine phosphatases (PTPs) such as PTP1B and SHP1 (5, 20). An established substrate of both Sts-1 and Sts-2, of 1 1 M (20, 26, 27). To identify small molecule inhibitors of Sts-1 for use as functional probes or like a basis for further drug development we miniaturized an established phosphatase assay and conducted a 20,000 compound high throughput screen of the Sts-1 phosphatase domain Sts-1HP. The screen yielded 51 active compounds (IC50 < 10 M) that were inactive in an enzyme-minus counter-screen. Within these hits were several groups of compounds with similar structural scaffolds. Among these, tetracycline analogs and sulfonate-containing azo dyes, were the largest two groups, with more than 10 and 5 variants present in the screening results, respectively. Both compound classes were determined to be competitive inhibitors of Sts-1HP that selectively inhibit this phosphatase, when compared to the PTPs SHP1 and PTP1B. Based on the conserved planar, aromatic structures of those inhibitors we speculated that aromatic residues within the protein were likely involved in critical protein-inhibitor interactions. Consistent with this hypothesis, mutation of either a tyrosine or tryptophan residue near the active site of Sts-1 reduces the effectiveness of the inhibitors without altering the kinetic parameters from the enzyme. Taken together, these data confirm the druggability from the Sts-1 phosphatase active site and offer the first types of selective Sts-1 inhibitors. The total results from the screen, aswell as the original system and structure-activity of actions data, give a foundation for future drug development efforts. DISCUSSION and RESULTS Assay design, miniaturization and testing: The Sts proteins, sts-1 particularly, have recently emerged as potential targets for the treating deadly pathogen infections (6C9). An altered immune response and increased fungal or bacterial clearance in Sts knockout mice generates a profound resistance phenotype (6C8, 28, 29). Small molecules that can and selectively inhibit Sts-1 phosphatase activity would be extremely valuable potently, both as probes of function and.[PMC free article] [PubMed] [Google Scholar] 48. display screen. Among the strikes had been two classes of structurally related substances, tetracycline variations and sulfonated azo dyes. These strikes acquired low micromolar to nanomolar IC50 beliefs. Orthogonal screening verified the validity of the inhibitors and showed that both action competitively on Sts-1 phosphatase activity. When examined on various other PTPs, PTP1B and SHP1, the tetracycline version, doxycycline, as well as the sulfonated azo dye, Congo crimson, are selective inhibitors of Sts-1Horsepower with selectivity indices which range from 19 to up to 200. The planar polyaromatic moieties within both classes of substances recommended a common binding setting. Mutation of either tryptophan 494 or tyrosine 596, located close to the energetic site from the proteins, reduced the from the inhibitors from 3- to 18-fold, indicating these residues can help promote binding of substrates with aromatic groupings. This function provides brand-new insights into substrate selectivity systems and represents two classes of substances that may serve as probes of function or being a basis for potential drug breakthrough. (6C9). This level of resistance phenotype is seen as a enhanced survival, speedy clearance from the pathogen and changed inflammatory response. Systemic attacks, with near 50,000 situations per year in america alone, have got high mortality prices that have not really declined within the last twenty years (10C12). mice claim that healing inactivation of Sts function generates a distinctive immune system response that really helps to decrease pathogen burden and stop the accompanying damaging inflammation. Therefore, the pharmacological inhibition of Sts activity presents a potential possibility to generate immuno-stimulatory therapies, to be utilized in conjunction with existing standard-of-care antibiotics or antifungals, as a way to treat dangerous pathogen attacks. Sts-1 and Sts-2, which talk about 40% sequence recognize, are multi-domain protein made up of an N-terminal ubiquitin association (UBA) domains, a Src-homology 3 (SH3) domains, and a C-terminal histidine phosphatase (HP) domain. The UBA and SH3 domains are thought to mediate protein-protein interactions and could be engaged in spatiotemporal regulation from the Sts catalytic function (16C18). The HP domain of the proteins are homologous towards the phosphoglycerate mutase (PGM) family, a subgroup from the histidine phosphatase superfamily (5, 19, 20). The HP domain from the Sts proteins, like other members from the PGM family, employs a 2-step catalytic mechanism that involves nucleophilic attack by one of the two conserved histidine residues and formation of a phosphor-histidine intermediate (19, 21C24). While the Sts proteins are known to catalyze the dephosphorylation of phosphotyrosines on protein substrates, they may be structurally, functionally and mechanistically distinct from canonical protein tyrosine phosphatases (PTPs) such as PTP1B and SHP1 (5, 20). An established substrate of both Sts-1 and Sts-2, of 1 1 M (20, 26, 27). To identify small molecule inhibitors of Sts-1 for use as functional probes or like a basis for further drug development we miniaturized an established phosphatase assay and conducted a 20,000 compound high throughput screen of the Sts-1 phosphatase domain Sts-1HP. The screen yielded 51 active compounds (IC50 < 10 M) that were inactive in an enzyme-minus counter-screen. Within these hits were several groups of Isorhamnetin 3-O-beta-D-Glucoside compounds with similar structural scaffolds. Among these, tetracycline analogs and sulfonate-containing azo dyes, were the largest two groups, with more than 10 and 5 variants present in the screening results, respectively. Both compound classes were determined to be competitive Isorhamnetin 3-O-beta-D-Glucoside inhibitors of Sts-1HP that selectively inhibit this phosphatase, when compared to the PTPs SHP1 and PTP1B. Based on the conserved planar, aromatic structures of these inhibitors we speculated that aromatic residues within the protein were likely involved in critical protein-inhibitor interactions. Consistent with this hypothesis, mutation of either a tyrosine or tryptophan residue near the active site of Sts-1 reduces the effectiveness of these inhibitors without altering.[PMC free article] [PubMed] [Google Scholar] 24. azo dyes. These hits experienced low micromolar to nanomolar IC50 ideals. Orthogonal screening confirmed the validity of these inhibitors and shown that both take action competitively on Sts-1 phosphatase activity. When tested on additional PTPs, PTP1B and SHP1, the tetracycline variant, doxycycline, and the sulfonated azo dye, Congo reddish, are selective inhibitors of Sts-1HP with selectivity indices ranging from 19 to as high as 200. The planar polyaromatic moieties present in both classes of compounds suggested a common binding mode. Mutation of either tryptophan 494 or tyrosine 596, located near the active site of the protein, reduced the of the inhibitors from 3- to 18-fold, indicating that these residues may help promote binding of substrates with aromatic organizations. This work provides fresh insights into substrate selectivity mechanisms and explains two classes of compounds that can serve as probes of function or like a basis for future drug finding. (6C9). This resistance phenotype is characterized by enhanced survival, quick clearance of the pathogen and modified inflammatory response. Systemic infections, with close to 50,000 instances per year in the United States alone, possess high mortality rates that have not declined in the last 20 years (10C12). mice suggest that restorative inactivation of Sts function generates a unique immune response that helps to reduce pathogen burden and prevent the accompanying harmful inflammation. As such, the pharmacological inhibition of Sts activity presents a potential opportunity to generate immuno-stimulatory therapies, to be used in combination with existing standard-of-care antibiotics or antifungals, as a means to treat fatal pathogen infections. Sts-1 and Sts-2, which share 40% sequence determine, are multi-domain proteins composed of an N-terminal ubiquitin association (UBA) website, a Src-homology 3 (SH3) website, and a C-terminal histidine phosphatase (HP) domain. The UBA and SH3 domains are believed to mediate protein-protein interactions and may be involved in spatiotemporal regulation of the Sts catalytic function (16C18). The HP domain of those proteins are homologous to the phosphoglycerate mutase (PGM) family, a subgroup of the histidine phosphatase superfamily (5, 19, 20). The HP domain of the Sts proteins, like other members of the PGM family, employs a 2-step catalytic mechanism that involves nucleophilic attack by one of the two conserved histidine residues and formation of a phosphor-histidine intermediate (19, 21C24). While the Sts proteins are known to catalyze the dephosphorylation of phosphotyrosines on protein substrates, they may be structurally, functionally and mechanistically distinct from canonical protein tyrosine phosphatases (PTPs) such as PTP1B and SHP1 (5, 20). An established substrate of both Sts-1 and Sts-2, of 1 1 M (20, 26, 27). To identify small molecule inhibitors of Sts-1 for use as functional probes or like a basis for further drug development we miniaturized an established phosphatase assay and conducted a 20,000 compound high throughput screen of the Sts-1 phosphatase domain Sts-1HP. The screen yielded 51 active compounds (IC50 < 10 M) that were inactive in an enzyme-minus counter-screen. Within these hits were several groups of compounds with similar structural scaffolds. Among these, tetracycline analogs and sulfonate-containing azo dyes, were the largest two groups, with more than 10 and 5 variants present in the screening results, respectively. Both compound classes were determined to be competitive inhibitors of Sts-1HP that selectively inhibit this phosphatase, when compared to the PTPs SHP1 and PTP1B. Based on the conserved planar, aromatic structures of those inhibitors we speculated that aromatic residues within the protein were likely involved in critical protein-inhibitor interactions. Consistent with this hypothesis, mutation of either a tyrosine or tryptophan residue near the active site of Sts-1 reduces the effectiveness of the inhibitors without altering the kinetic parameters from the enzyme. Taken together, the druggability is confirmed by these data of the Sts-1 phosphatase active site.[PMC free article] [PubMed] [Google Scholar] 50. Hereditary inactivation from the Sts protein dramatically increases web host success of systemic infections and qualified prospects to improved pathogen clearance. The proteins tyrosine phosphatase (PTP) activity of the proteins comes from a C-terminal 2-histidine phosphatase (Horsepower) domain. To recognize new inhibitors from the Horsepower activity of Sts-1 we miniaturized a phosphatase assay to at least one 1,536-well format and executed a 20,580 substance display screen. Among the strikes had been two classes of structurally related substances, tetracycline variations and sulfonated azo dyes. These strikes got low micromolar to nanomolar IC50 beliefs. Orthogonal screening verified the validity of the inhibitors and confirmed that both work competitively on Sts-1 phosphatase activity. When examined on various other PTPs, PTP1B and SHP1, the tetracycline version, doxycycline, as well as the sulfonated azo dye, Congo reddish colored, are selective inhibitors of Sts-1Horsepower with selectivity indices which range from 19 to up to 200. The planar polyaromatic moieties within both classes of substances recommended a common binding setting. Mutation of either tryptophan 494 or tyrosine 596, located close to the energetic site from the proteins, reduced the from the inhibitors from 3- to 18-fold, indicating these residues can help promote binding of substrates with aromatic groupings. This function provides brand-new insights into substrate selectivity systems and details two classes of substances that may serve as probes of function or being a basis for future drug discovery. (6C9). This resistance phenotype is seen as a enhanced survival, rapid clearance from the pathogen and altered inflammatory response. Systemic infections, with near 50,000 cases each year in america alone, have high mortality rates which have not declined within the last twenty years (10C12). mice claim that therapeutic inactivation of Sts function generates a distinctive immune response that really helps to reduce pathogen burden and stop the accompanying destructive inflammation. Therefore, the pharmacological inhibition of Sts activity presents a potential possibility to generate immuno-stimulatory therapies, to be utilized in conjunction with existing standard-of-care antibiotics or antifungals, as a way to take care of deadly pathogen infections. Sts-1 and Sts-2, which share 40% sequence identify, are multi-domain proteins made up of an N-terminal ubiquitin association (UBA) domain, a Src-homology 3 (SH3) domain, and a C-terminal histidine phosphatase (HP) domain. The UBA and SH3 domains are thought to mediate protein-protein interactions and could be engaged in spatiotemporal regulation from the Sts catalytic function (16C18). The HP domain of the proteins are homologous towards the phosphoglycerate mutase (PGM) family, a subgroup from the histidine phosphatase superfamily (5, 19, 20). The HP domain from the Sts proteins, like other members from the PGM family, employs a 2-step catalytic mechanism which involves nucleophilic attack by among the two conserved histidine residues and formation of the phosphor-histidine intermediate (19, 21C24). As the Sts proteins are recognized to catalyze the dephosphorylation of phosphotyrosines on protein substrates, these are structurally, functionally and mechanistically distinct from canonical protein tyrosine phosphatases (PTPs) such as for example PTP1B and SHP1 (5, 20). A recognised substrate of both Sts-1 and Sts-2, of just one 1 M (20, 26, 27). To recognize small molecule inhibitors of Sts-1 for use as functional probes or being a basis for even more drug development we miniaturized a recognised phosphatase assay and conducted a 20,000 compound high throughput screen from the Sts-1 phosphatase domain Sts-1HP. The screen yielded 51 active compounds (IC50 < 10 M) which were inactive within an enzyme-minus counter-screen. Within these hits were several sets of compounds with similar structural scaffolds. Among these, tetracycline analogs and sulfonate-containing azo dyes, were the biggest two groups, with an increase of than 10 and 5 variants within the screening results, respectively. Both compound classes were determined to compete inhibitors of Sts-1HP that selectively inhibit this phosphatase, in comparison with the PTPs SHP1 and PTP1B. Predicated on the conserved planar, aromatic structures of the inhibitors we speculated that aromatic residues for the protein were likely involved with critical protein-inhibitor.