Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
J Transl Autoimmun ; 6: 100185, 2023.
Article in English | MEDLINE | ID: mdl-36654851

ABSTRACT

Dual-specificity tyrosine phosphorylation-regulated kinase 1 A (DYRK1A) has been proposed as a novel regulator of adaptive immune homeostasis through modulating T cell polarization. Thus, DYRK1A could present a potential target in autoimmune disorders. Here, we identify FRTX-02 as a novel compound exhibiting potent and selective inhibition of DYRK1A. FRTX-02 induced transcriptional activity of the DYRK1A substrate NFAT in T cell lines. Correspondingly, FRTX-02 promoted ex vivo CD4+ polarization into anti-inflammatory Tregs and reduced their polarization into pro-inflammatory Th1 or Th17 cells. We show that FRTX-02 could also limit innate immune responses through negative regulation of the MyD88/IRAK4-NF-κB axis in a mast cell line. Finally, in mouse models of psoriasis and atopic dermatitis, both oral and topical formulations of FRTX-02 reduced inflammation and disease biomarkers in a dose-dependent manner. These results support further studies of DYRK1A inhibitors, including FRTX-02, as potential therapies for chronic inflammatory and autoimmune conditions.

2.
J Med Chem ; 64(10): 6985-6995, 2021 05 27.
Article in English | MEDLINE | ID: mdl-33942608

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive breast-cancer subtype associated with poor prognosis and high relapse rates. Monopolar spindle 1 kinase (MPS1) is an apical dual-specificity protein kinase that is over-expressed in TNBC. We herein report a highly selective MPS1 inhibitor based on a 7H-pyrrolo[2,3-d]pyrimidine-5-carbonitrile scaffold. Our lead optimization was guided by key X-ray crystal structure analysis. In vivo evaluation of candidate (9) is shown to effectively mitigate human TNBC cell proliferation.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Drug Design , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/chemistry , Pyrroles/chemistry , Administration, Oral , Animals , Binding Sites , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Crystallography, X-Ray , Female , Half-Life , Humans , Mice , Mice, Inbred ICR , Molecular Docking Simulation , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Pyrimidines/metabolism , Pyrimidines/therapeutic use , Pyrroles/metabolism , Pyrroles/therapeutic use , Structure-Activity Relationship , Transplantation, Heterologous
3.
Cell Death Dis ; 9(11): 1125, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30420654

ABSTRACT

Leucine-rich repeat kinase 2 (LRRK2), originally identified as a causative genetic factor in Parkinson's disease, is now associated with a number of pathologies. Here, we show that brain injury induces a robust expression of endogenous LRRK2 and suggest a role of LRRK2 after injury. We found that various in vitro and in vivo models of traumatic brain injury (TBI) markedly enhanced LRRK2 expression in neurons and also increased the level of hypoxia-inducible factor (HIF)-1α. Luciferase reporter assay and chromatin immunoprecipitation revealed direct binding of HIF-1α in LRRK2 proximal promoter. We also found that HIF-1α-dependent transcriptional induction of LRRK2 exacerbated neuronal cell death following injury. Furthermore, application of G1023, a specific, brain-permeable inhibitor of LRRK2, substantially prevented brain tissue damage, cell death, and inflammatory response and alleviated motor and cognitive defects induced by controlled cortical impact injury. Together, these results suggest HIF-1α-LRRK2 axis as a potential therapeutic target for brain injury.


Subject(s)
Brain Injuries, Traumatic/genetics , Cerebral Cortex/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Transcription, Genetic , Animals , Apoptosis/drug effects , Apoptosis/genetics , Base Sequence , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/prevention & control , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Female , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/antagonists & inhibitors , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Mice , Mice, Inbred ICR , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , Protein Kinase Inhibitors/pharmacology , Psychomotor Performance/drug effects , Signal Transduction
4.
J Neuroinflammation ; 15(1): 286, 2018 Oct 11.
Article in English | MEDLINE | ID: mdl-30309372

ABSTRACT

BACKGROUND: Neuroinflammation is associated with neurodegenerative diseases, including Alzheimer's disease (AD). Thus, modulating the neuroinflammatory response represents a potential therapeutic strategy for treating neurodegenerative diseases. Several recent studies have shown that dopamine (DA) and its receptors are expressed in immune cells and are involved in the neuroinflammatory response. Thus, we recently developed and synthesized a non-self-polymerizing analog of DA (CA140) and examined the effect of CA140 on neuroinflammation. METHODS: To determine the effects of CA140 on the neuroinflammatory response, BV2 microglial cells were pretreated with lipopolysaccharide (LPS, 1 µg/mL), followed by treatment with CA140 (10 µM) and analysis by reverse transcription-polymerase chain reaction (RT-PCR). To examine whether CA140 alters the neuroinflammatory response in vivo, wild-type mice were injected with both LPS (10 mg/kg, intraperitoneally (i.p.)) and CA140 (30 mg/kg, i.p.), and immunohistochemistry was performed. In addition, familial AD (5xFAD) mice were injected with CA140 or vehicle daily for 2 weeks and examined for microglial and astrocyte activation. RESULTS: Pre- or post-treatment with CA140 differentially regulated proinflammatory responses in LPS-stimulated microglia and astrocytes. Interestingly, CA140 regulated D1R levels to alter LPS-induced proinflammatory responses. CA140 significantly downregulated LPS-induced phosphorylation of ERK and STAT3 in BV2 microglia cells. In addition, CA140-injected wild-type mice exhibited significantly decreased LPS-induced microglial and astrocyte activation. Moreover, CA140-injected 5xFAD mice exhibited significantly reduced microglial and astrocyte activation. CONCLUSIONS: CA140 may be beneficial for preventing and treating neuroinflammatory-related diseases, including AD.


Subject(s)
Alzheimer Disease/complications , Anti-Inflammatory Agents/therapeutic use , Dopamine/analogs & derivatives , Encephalitis/drug therapy , Encephalitis/etiology , Alzheimer Disease/blood , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/genetics , Animals , Animals, Newborn , Brain/metabolism , Brain/pathology , Cells, Cultured , Disease Models, Animal , Dopamine/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Mutation/genetics , Nerve Tissue Proteins/metabolism , Polysaccharides/pharmacology , Presenilin-1/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
5.
Cell Chem Biol ; 25(1): 88-99.e6, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29129717

ABSTRACT

Heterobifunctional molecules that recruit E3 ubiquitin ligases, such as cereblon, for targeted protein degradation represent an emerging pharmacological strategy. A major unanswered question is how generally applicable this strategy is to all protein targets. In this study, we designed a multi-kinase degrader by conjugating a highly promiscuous kinase inhibitor with a cereblon-binding ligand, and used quantitative proteomics to discover 28 kinases, including BTK, PTK2, PTK2B, FLT3, AURKA, AURKB, TEC, ULK1, ITK, and nine members of the CDK family, as degradable. This set of kinases is only a fraction of the intracellular targets bound by the degrader, demonstrating that successful degradation requires more than target engagement. The results guided us to develop selective degraders for FLT3 and BTK, with potentials to improve disease treatment. Together, this study demonstrates an efficient approach to triage a gene family of interest to identify readily degradable targets for further studies and pre-clinical developments.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proteomics , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase/genetics , Agammaglobulinaemia Tyrosine Kinase/metabolism , Humans , Protein Kinase Inhibitors/chemistry , Proteolysis , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
6.
J Med Chem ; 60(17): 7569-7578, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28816449

ABSTRACT

Although Hsp90 inhibitors can inhibit multiple tumorigenic pathways in cancer cells, their anticancer activity has been disappointingly modest. However, by forcing Hsp90 inhibitors into the mitochondria with mitochondrial delivery vehicles, they were converted into potent drugs targeting the mitochondrial Hsp90 paralog TRAP1. Here, to improve mitochondrial drug accumulation without using the mitochondrial delivery vehicle, we increased freely available drug concentrations in the cytoplasm by reducing the binding of the drugs to the abundant cytoplasmic Hsp90. After analyzing X-ray cocrystal structures, the purine ring of the Hsp90 inhibitor 2 (BIIB021) was modified to pyrazolopyrimidine scaffolds. One pyrazolopyrimidine, 12b (DN401), bound better to TRAP1 than to Hsp90, inactivated the mitochondrial TRAP1 in vivo, and it exhibited potent anticancer activity. Therefore, the rationale and feasible guidelines for developing 12b can potentially be exploited to design a potent TRAP1 inhibitor.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cell Death/drug effects , Cell Line, Tumor , Crystallography, X-Ray , HSP90 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Molecular Docking Simulation , Neoplasms/metabolism , Neoplasms/pathology , Pyrazoles/pharmacokinetics , Pyrazoles/therapeutic use , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use
7.
ACS Chem Biol ; 12(10): 2570-2578, 2017 10 20.
Article in English | MEDLINE | ID: mdl-28767222

ABSTRACT

Proteolysis targeting chimera (PROTAC) technology, the recruitment of E3 ubiquitin ligases to induce the degradation of a protein target, is rapidly impacting chemical biology, as well as modern drug development. Here, we explore the universality of this approach by evaluating different E3 ubiquitin ligases, engineered in their substrate binding domains to accept a recruiting ligand. Five out of six E3 ligases were found to be amenable to recruitment for target degradation. Taking advantage of the tight spatiotemporal control of inducing ubiquitination on a preselected target in living cells, we focused on two of the engineered E3 ligases, ßTRCP and parkin, to unravel their ubiquitination characteristics in comparison with the PROTAC-recruited endogenous E3 ligases VHL and cereblon.


Subject(s)
Ubiquitin-Protein Ligases/metabolism , Cell Line , Cloning, Molecular , Humans , Models, Molecular , Molecular Structure , Protein Conformation , Proteolysis , Ubiquitin-Protein Ligases/genetics , Ubiquitination/drug effects
8.
Oncotarget ; 7(50): 83308-83318, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27829217

ABSTRACT

Breast cancer is the most common malignant disease occurring in women and represents a substantial proportion of the global cancer burden. In these patients, metastasis but not the primary tumor is the main cause of breast cancer-related deaths. Here, we report the novel finding that DN10764 (AZD7762, a selective inhibitor of checkpoint kinases 1 and 2) can suppress breast cancer metastasis. In breast cancer cells, DN10764 inhibited cell proliferation and GAS6-mediated AXL signaling, consequently resulting in suppressed migration and invasion. In addition, DN10764 induced caspase 3/7-mediated apoptosis in breast cancer cells and inhibited tube formation of human umbilical vein endothelial cells. Finally, DN10764 significantly suppressed the tumor growth and metastasis of breast cancer cells in in vivo metastasis models. Taken together, these data suggest that therapeutic strategies targeting AXL in combination with systemic therapies could improve responses to anti-cancer therapies and reduce breast cancer recurrence and metastases.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Movement/drug effects , Lung Neoplasms/prevention & control , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction/drug effects , Thiophenes/pharmacology , Urea/analogs & derivatives , A549 Cells , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/secondary , Animals , Apoptosis/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspase 3/metabolism , Caspase 7/metabolism , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Human Umbilical Vein Endothelial Cells/pathology , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Physiologic/drug effects , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Time Factors , Transfection , Tumor Burden/drug effects , Urea/pharmacology , Xenograft Model Antitumor Assays , Axl Receptor Tyrosine Kinase
9.
Angew Chem Int Ed Engl ; 55(2): 807-10, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26593377

ABSTRACT

Proteolysis Targeting Chimera (PROTAC) technology is a rapidly emerging alternative therapeutic strategy with the potential to address many of the challenges currently faced in modern drug development programs. PROTAC technology employs small molecules that recruit target proteins for ubiquitination and removal by the proteasome. The synthesis of PROTAC compounds that mediate the degradation of c-ABL and BCR-ABL by recruiting either Cereblon or Von Hippel Lindau E3 ligases is reported. During the course of their development, we discovered that the capacity of a PROTAC to induce degradation involves more than just target binding: the identity of the inhibitor warhead and the recruited E3 ligase largely determine the degradation profiles of the compounds; thus, as a starting point for PROTAC development, both the target ligand and the recruited E3 ligase should be varied to rapidly generate a PROTAC with the desired degradation profile.


Subject(s)
Fusion Proteins, bcr-abl/metabolism , Cell Line , Cell Line, Tumor , Humans , Proteolysis
10.
Nat Chem Biol ; 11(8): 611-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26075522

ABSTRACT

The current predominant therapeutic paradigm is based on maximizing drug-receptor occupancy to achieve clinical benefit. This strategy, however, generally requires excessive drug concentrations to ensure sufficient occupancy, often leading to adverse side effects. Here, we describe major improvements to the proteolysis targeting chimeras (PROTACs) method, a chemical knockdown strategy in which a heterobifunctional molecule recruits a specific protein target to an E3 ubiquitin ligase, resulting in the target's ubiquitination and degradation. These compounds behave catalytically in their ability to induce the ubiquitination of super-stoichiometric quantities of proteins, providing efficacy that is not limited by equilibrium occupancy. We present two PROTACs that are capable of specifically reducing protein levels by >90% at nanomolar concentrations. In addition, mouse studies indicate that they provide broad tissue distribution and knockdown of the targeted protein in tumor xenografts. Together, these data demonstrate a protein knockdown system combining many of the favorable properties of small-molecule agents with the potent protein knockdown of RNAi and CRISPR.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinase 2/antagonists & inhibitors , Receptors, Estrogen/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Binding Sites , Biocatalysis , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Humans , MCF-7 Cells , Mice , Models, Molecular , Molecular Targeted Therapy , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Proteolysis , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitination , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , ERRalpha Estrogen-Related Receptor
11.
PLoS One ; 9(3): e89617, 2014.
Article in English | MEDLINE | ID: mdl-24603864

ABSTRACT

A library of peptidomimetics was assembled combinatorially into dimers on a triazine-based core. The pharmacophore corresponds to ß-turns of the neurotrophin polypeptides neurotrophin-3 (NT-3), nerve growth factor (NGF), or brain-derived neurotrophic factor (BDNF). These are the natural ligands for TrkC, TrkA, and TrkB receptors, respectively. The linker length and the side-chain orientation of each monomer within the bivalent mimics were systematically altered, and the impact of these changes on the function of each ligand was evaluated. While the monovalent peptidomimetics had no detectable binding or bioactivity, four bivalent peptidomimetics (2c, 2d, 2e, 3f) are selective TrkC ligands with antagonistic activity, and two bivalent peptidomimetics (1a, 1b) are TrkC and TrkA ligands with antagonistic activity. All these bivalent compounds block ligand-dependent receptor activation and cell survival, without affecting neuritogenic differentiation. This work adds to our understanding of how the neurotrophins function through Trk receptors, and demonstrates that peptidomimetics can be designed to selectively disturb specific biological signals, and may be used as pharmacological probes or as therapeutic leads. The concept of altering side-chain, linker length, and sequence orientation of a subunit within a pharmacophore provides an easy modular approach to generate larger libraries with diversified bioactivity.


Subject(s)
Combinatorial Chemistry Techniques/methods , Peptidomimetics/pharmacology , Receptor, trkA/antagonists & inhibitors , Receptor, trkC/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Blotting, Western , Brain-Derived Neurotrophic Factor/chemistry , Brain-Derived Neurotrophic Factor/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Models, Chemical , Molecular Structure , NIH 3T3 Cells , Nerve Growth Factor/chemistry , Nerve Growth Factor/pharmacology , Neurotrophin 3/chemistry , Neurotrophin 3/pharmacology , PC12 Cells , Peptidomimetics/chemical synthesis , Peptidomimetics/chemistry , Rats , Receptor, trkA/genetics , Receptor, trkA/metabolism , Receptor, trkC/genetics , Receptor, trkC/metabolism , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry
12.
Org Biomol Chem ; 11(44): 7789-801, 2013 Nov 28.
Article in English | MEDLINE | ID: mdl-24121516

ABSTRACT

Peptide mimics that display amino acid side-chains on semi-rigid scaffolds (not peptide polyamides) can be referred to as minimalist mimics. Accessible conformations of these scaffolds may overlay with secondary structures giving, for example, "minimalist helical mimics". It is difficult for researchers who want to apply minimalist mimics to decide which one to use because there is no widely accepted protocol for calibrating how closely these compounds mimic secondary structures. Moreover, it is also difficult for potential practitioners to evaluate which ideal minimalist helical mimics are preferred for a particular set of side-chains. For instance, what mimic presents i, i + 4, i + 7 side-chains in orientations that best resemble an ideal α-helix, and is a different mimic required for a i, i + 3, i + 7 helical combination? This article describes a protocol for fitting each member of an array of accessible scaffold conformations on secondary structures. The protocol involves: (i) use quenched molecular dynamics (QMD) to generate an ensemble consisting of hundreds of accessible, low energy conformers of the mimics; (ii) representation of each of these as a set of Cα and Cß coordinates corresponding to three amino acid side-chains displayed by the scaffolds; (iii) similar representation of each combination of three side-chains in each ideal secondary structure as a set of Cα and Cß coordinates corresponding to three amino acid side-chains displayed by the scaffolds; and, (iv) overlay Cα and Cß coordinates of all the conformers on all the sets of side-chain "triads" in the ideal secondary structures and express the goodness of fit in terms of root mean squared deviation (RMSD, Å) for each overlay. We refer to this process as Exploring Key Orientations on Secondary structures (EKOS). Application of this procedure reveals the relative bias of a scaffold to overlay on different secondary structures, the "side-chain correspondences" (e.g. i, i + 4, i + 7 or i, i + 3, i + 4) of those overlays, and the energy of this state relative to the minimum located. This protocol was tested on some of the most widely cited minimalist α-helical mimics (1-8 in the text). The data obtained indicates several of these compounds preferentially exist in conformations that resemble other secondary structures as well as α-helices, and many of the α-helical conformations have unexpected side-chain correspondences. These observations imply the featured minimalist mimics have more scope for disrupting PPI interfaces than previously anticipated. Finally, the same simulation method was used to match preferred conformations of minimalist mimics with actual protein/peptide structures at interfaces providing quantitative comparisons of predicted fits of the test mimics at protein-protein interaction sites.


Subject(s)
Molecular Mimicry , Protein Structure, Secondary
13.
J Am Chem Soc ; 135(1): 167-73, 2013 Jan 09.
Article in English | MEDLINE | ID: mdl-23270593

ABSTRACT

Small molecule probes that selectively perturb protein-protein interactions (PPIs) are pivotal to biomedical science, but their discovery is challenging. We hypothesized that conformational resemblance of semirigid scaffolds expressing amino acid side-chains to PPI-interface regions could guide this process. Consequently, a data mining algorithm was developed to sample huge numbers of PPIs to find ones that match preferred conformers of a selected semirigid scaffold. Conformations of one such chemotype (1aaa; all methyl side-chains) matched several biomedically significant PPIs, including the dimerization interface of HIV-1 protease. On the basis of these observations, four molecules 1 with side-chains corresponding to the matching HIV-1 dimerization interface regions were prepared; all four inhibited HIV-1 protease via perturbation of dimerization. These data indicate this approach may inspire design of small molecule interface probes to perturb PPIs.


Subject(s)
Fluorescent Dyes/pharmacology , HIV Protease/chemistry , Small Molecule Libraries/pharmacology , Algorithms , Dimerization , Fluorescent Dyes/chemistry , HIV Protease/metabolism , Models, Molecular , Protein Binding/drug effects , Small Molecule Libraries/chemistry , Structure-Activity Relationship
14.
ACS Med Chem Lett ; 3(12): 1008-1012, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-23411915

ABSTRACT

A small molecule motif was used in "active targeting" to deliver cytotoxic substances into tumor cells that express the TrkC receptor. Underlying this study was the hypothesis that internalization of targeted conjugates into cells would be facile if mediated by receptor binding and receptor-ligand internalization. Initial experiments using 6-mercaptopurine gave encouraging data, but demonstrated the importance of maintaining solubility and high cytotoxicity. Conjugates of the targeting agent with a cytotoxic rosamine (similar to a rhodamine) were more successful. Targeting of TrkC was observed, validated in a series of competition experiments featuring other TrkC ligands, and accumulation into lysosomes was observed, as expected for receptor-mediated internalization.

16.
J Am Chem Soc ; 133(32): 12350-3, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21780756

ABSTRACT

Peptidomimetics 1-3 were prepared from amino acid-derived tetramic acids 7 as the key starting materials. Calculations show that preferred conformations of 1 can align their side-chain vectors with amino acids in common secondary structures more effectively than conformations of 3. A good fit was found for a preferred conformation of 2 (an extended derivative of 1) with a sheet/ß-turn/sheet motif.


Subject(s)
Peptidomimetics/chemistry , Pyrroles/chemistry , Pyrrolidines/chemistry , Models, Molecular , Peptidomimetics/chemical synthesis , Protein Structure, Secondary , Pyrroles/chemical synthesis , Pyrrolidines/chemical synthesis
17.
Chem Soc Rev ; 40(8): 4411-21, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21483946

ABSTRACT

Many "new generation" peptidomimetics are designed to present amino acid side chains only; they do not have structural features that resemble peptide main chains. These types of molecules have frequently been presented in the literature as mimics of specific secondary structures. However, many "side-chain only" peptidomimetics do not rest in single conformational states, but exist in a limited number of freely interconverting forms. These different conformations may resemble different secondary structures, so referring to them as, for instance, turn- or helical-mimics understates the ways they could adapt to various binding situations. Sets of scaffolds that can be used to mimic aspects of nearly every secondary structure, i.e. universal peptidomimetics, can be constructed. These may assume a privileged place in library design, particularly in high throughput screening for pharmacological probes for which binding conformations, or even the target itself, is unknown at the time the library is designed (critical review, 101 references).


Subject(s)
Peptidomimetics , Models, Molecular , Molecular Structure , Peptides/chemistry
18.
Org Lett ; 13(5): 980-3, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21268601

ABSTRACT

Two amino acid derived synthons were combined to give homopropargylic amines 2. Platinum dichloride was used to cyclize these intermediates into pyrroles 3 which collapsed to the target secondary structure mimics 1 on treatment with base. Side chains of these compounds overlay with an idealized type 1 ß-turn and with an inverse γ-turn.


Subject(s)
Amines/chemical synthesis , Amino Acids/chemistry , Pyrroles/chemistry , Amines/chemistry , Combinatorial Chemistry Techniques , Cyclization , Models, Molecular , Molecular Structure , Peptidomimetics , Protein Structure, Secondary
19.
J Am Chem Soc ; 133(3): 462-77, 2011 Jan 26.
Article in English | MEDLINE | ID: mdl-21182254

ABSTRACT

This paper concerns peptidomimetic scaffolds that can present side chains in conformations resembling those of amino acids in secondary structures without incurring excessive entropic or enthalpic penalties. Compounds of this type are referred to here as minimalist mimics. The core hypothesis of this paper is that small sets of such scaffolds can be designed to analogue local pairs of amino acids (including noncontiguous ones) in any secondary structure; i.e., they are universal peptidomimetics. To illustrate this concept, we designed a set of four peptidomimetic scaffolds. Libraries based on them were made bearing side chains corresponding to many of the protein-derived amino acids. Modeling experiments were performed to give an indication of kinetic and thermodynamic accessibilities of conformations that can mimic secondary structures. Together, peptidomimetics based on these four scaffolds can adopt conformations that resemble almost any combination of local amino acid side chains in any secondary structure. Universal peptidomimetics of this kind are likely to be most useful in the design of libraries for high-throughput screening against diverse targets. Consequently, data arising from submission of these molecules to the NIH Molecular Libraries Small Molecule Repository (MLSMR) are outlined.


Subject(s)
Peptidomimetics , Kinetics , Models, Molecular , Protein Structure, Secondary , Quantum Theory , Thermodynamics
20.
Invest Ophthalmol Vis Sci ; 51(12): 6639-51, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20574020

ABSTRACT

PURPOSE: Glaucoma is a distinct neuropathy characterized by the chronic and progressive death of retinal ganglion cells (RGCs). The etiology of RGC death remains unknown. Risk factors for glaucomatous RGC death are elevated intraocular pressure and glial production of tumor necrosis factor-alpha (TNF-α). Previously, the authors showed that glaucoma causes a rapid upregulation of a neurotrophin receptor truncated isoform lacking the kinase domain, TrkC.T1, in retina. Here they examined the biological role of TrkC.T1 during glaucoma progression. METHODS: Rat and mouse models of chronic ocular hypertension were used. Immunofluorescence Western blot analysis and in situ mRNA hybridization were used to identify cells upregulating TrkC.T1. A genetic model of engineered mice lacking TrkC.T1 (TrkC.T1(-/-)) was used to validate a role for this receptor in glaucoma. Pharmacologic studies were conducted to evaluate intravitreal delivery of agonists or antagonists of TrkC.T1, compared with controls, during glaucoma. Surviving RGCs were quantified by retrograde-labeling techniques. Production of neurotoxic TNF-α and α2 macroglobulin were quantified. RESULTS: TrkC.T1 was upregulated in retinal glia, with a pattern similar to that of TNF-α. TrkC.T1(-/-) mice had normal retinas. However, during experimental glaucoma, TrkC.T1(-/-) mice had lower rates of RGC death and produced less TNF-α than wild-type littermates. In rats with glaucoma, the pharmacologic use of TrkC antagonists delayed RGC death and reduced the production of retinal TNF-α. CONCLUSIONS: TrkC.T1 is implicated in glaucomatous RGC death through the control of glial TNF-α production. Overall, the data point to a paracrine mechanism whereby elevated intraocular pressure upregulated glial TrkC.T1 expression in glia; TrkC.T1 controlled glial TNF-α production, and TNF-α caused RGC death.


Subject(s)
Disease Models, Animal , Glaucoma/metabolism , Neuroglia/metabolism , Receptor, trkC/metabolism , Retinal Ganglion Cells/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Blotting, Western , Cell Death , Electroporation , Female , Fluorescent Antibody Technique, Indirect , Genetic Vectors , In Situ Hybridization , Intraocular Pressure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Isoforms/agonists , Protein Isoforms/analysis , Protein Isoforms/metabolism , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptor, trkC/agonists , Receptor, trkC/antagonists & inhibitors , Up-Regulation , alpha-Macroglobulins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...