37 results on '"Backes, BJ"'
Search Results
2. Targeting ABL-IRE1 alpha Signaling Spares ER-Stressed Pancreatic beta Cells to Reverse Autoimmune Diabetes (vol 25, pg 883, 2017)
- Author
-
Morita, S, Villalta, SA, Feldman, HC, Register, AC, Rosenthal, W, Hoffmann-Petersen, IT, Mehdizadeh, M, Ghosh, R, Wang, L, Colon-Negron, K, Meza-Acevedo, R, Backes, BJ, Maly, DJ, Bluestone, JA, and Papa, FR
- Published
- 2017
3. Targeting ABL-IRE1 alpha Signaling Spares ER- Stressed Pancreatic beta Cells to Reverse Autoimmune Diabetes
- Author
-
Morita, S, Villalta, SA, Feldman, HC, Register, AC, Rosenthal, W, Hoffmann-Petersen, IT, Mehdizadeh, M, Ghosh, R, Wang, L, Colon-Negron, K, Meza-Acevedo, R, Backes, BJ, Maly, DJ, Bluestone, JA, and Papa, FR
- Published
- 2017
4. IRE1α drives lung epithelial progenitor dysfunction to establish a niche for pulmonary fibrosis.
- Author
-
Auyeung VC, Downey MS, Thamsen M, Wenger TA, Backes BJ, Sheppard D, and Papa FR
- Subjects
- Apoptosis physiology, Endoplasmic Reticulum Stress physiology, Humans, Lung metabolism, Protein Serine-Threonine Kinases genetics, Endoribonucleases metabolism, Idiopathic Pulmonary Fibrosis metabolism
- Abstract
After lung injury, damage-associated transient progenitors (DATPs) emerge, representing a transitional state between injured epithelial cells and newly regenerated alveoli. DATPs express profibrotic genes, suggesting that they might promote idiopathic pulmonary fibrosis (IPF). However, the molecular pathways that induce and/or maintain DATPs are incompletely understood. Here we show that the bifunctional kinase/RNase-IRE1α-a central mediator of the unfolded protein response (UPR) to endoplasmic reticulum (ER) stress is a critical promoter of DATP abundance and function. Administration of a nanomolar-potent, monoselective kinase inhibitor of IRE1α (KIRA8)-or conditional epithelial IRE1α gene knockout-both reduce DATP cell number and fibrosis in the bleomycin model, indicating that IRE1α cell-autonomously promotes transition into the DATP state. IRE1α enhances the profibrotic phenotype of DATPs since KIRA8 decreases expression of integrin αvβ6, a key activator of transforming growth factor β (TGF-β) in pulmonary fibrosis, corresponding to decreased TGF-β-induced gene expression in the epithelium and decreased collagen accumulation around DATPs. Furthermore, IRE1α regulates DNA damage response (DDR) signaling, previously shown to promote the DATP phenotype, as IRE1α loss-of-function decreases H2AX phosphorylation, Cdkn1a (p21) expression, and DDR-associated secretory gene expression. Finally, KIRA8 treatment increases the differentiation of Krt19
CreERT2 -lineage-traced DATPs into type 1 alveolar epithelial cells after bleomycin injury, indicating that relief from IRE1α signaling enables DATPs to exit the transitional state. Thus, IRE1α coordinates a network of stress pathways that conspire to entrap injured cells in the DATP state. Pharmacological blockade of IRE1α signaling helps resolve the DATP state, thereby ameliorating fibrosis and promoting salutary lung regeneration.- Published
- 2022
- Full Text
- View/download PDF
5. ATP-competitive partial antagonists of the IRE1α RNase segregate outputs of the UPR.
- Author
-
Feldman HC, Ghosh R, Auyeung VC, Mueller JL, Kim JH, Potter ZE, Vidadala VN, Perera BGK, Olivier A, Backes BJ, Zikherman J, Papa FR, and Maly DJ
- Subjects
- Adenosine Triphosphate chemistry, Endoribonucleases metabolism, Humans, Models, Molecular, Molecular Structure, Protein Kinase Inhibitors chemistry, Protein Serine-Threonine Kinases metabolism, Protein Unfolding drug effects, Adenosine Triphosphate pharmacology, Endoribonucleases antagonists & inhibitors, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
The unfolded protein response (UPR) homeostatically matches endoplasmic reticulum (ER) protein-folding capacity to cellular secretory needs. However, under high or chronic ER stress, the UPR triggers apoptosis. This cell fate dichotomy is promoted by differential activation of the ER transmembrane kinase/endoribonuclease (RNase) IRE1α. We previously found that the RNase of IRE1α can be either fully activated or inactivated by ATP-competitive kinase inhibitors. Here we developed kinase inhibitors, partial antagonists of IRE1α RNase (PAIRs), that partially antagonize the IRE1α RNase at full occupancy. Biochemical and structural studies show that PAIRs promote partial RNase antagonism by intermediately displacing the helix αC in the IRE1α kinase domain. In insulin-producing β-cells, PAIRs permit adaptive splicing of Xbp1 mRNA while quelling destructive ER mRNA endonucleolytic decay and apoptosis. By preserving Xbp1 mRNA splicing, PAIRs allow B cells to differentiate into immunoglobulin-producing plasma cells. Thus, an intermediate RNase-inhibitory 'sweet spot', achieved by PAIR-bound IRE1α, captures a desirable conformation for drugging this master UPR sensor/effector., (© 2021. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2021
- Full Text
- View/download PDF
6. A defect in COPI-mediated transport of STING causes immune dysregulation in COPA syndrome.
- Author
-
Deng Z, Chong Z, Law CS, Mukai K, Ho FO, Martinu T, Backes BJ, Eckalbar WL, Taguchi T, and Shum AK
- Subjects
- Animals, Endoplasmic Reticulum metabolism, Fibroblasts metabolism, Gene Knock-In Techniques, Golgi Apparatus metabolism, HEK293 Cells, Homeostasis immunology, Humans, Interferon Type I metabolism, Membrane Proteins genetics, Mice, Mice, Inbred C57BL, Mice, Transgenic, Mutation, Missense, Protein Transport genetics, Signal Transduction genetics, Syndrome, Transfection, Coatomer Protein genetics, Coatomer Protein metabolism, Immune System Diseases genetics, Membrane Proteins metabolism
- Abstract
Pathogenic COPA variants cause a Mendelian syndrome of immune dysregulation with elevated type I interferon signaling. COPA is a subunit of coat protein complex I (COPI) that mediates Golgi to ER transport. Missense mutations of the COPA WD40 domain impair binding and sorting of proteins targeted for ER retrieval, but how this causes disease remains unknown. Given the importance of COPA in Golgi-ER transport, we speculated that type I interferon signaling in COPA syndrome involves missorting of STING. We show that a defect in COPI transport causes ligand-independent activation of STING. Furthermore, SURF4 is an adapter molecule that facilitates COPA-mediated retrieval of STING at the Golgi. Activated STING stimulates type I interferon-driven inflammation in CopaE241K/+ mice that is rescued in STING-deficient animals. Our results demonstrate that COPA maintains immune homeostasis by regulating STING transport at the Golgi. In addition, activated STING contributes to immune dysregulation in COPA syndrome and may be a new molecular target in treating the disease., Competing Interests: Disclosures: T. Martinu reported grants from Sanofi and non-financial support from APCBio outside the submitted work. No other disclosures were reported., (© 2020 Deng et al.)
- Published
- 2020
- Full Text
- View/download PDF
7. Targeting Adaptive IRE1α Signaling and PLK2 in Multiple Myeloma: Possible Anti-Tumor Mechanisms of KIRA8 and Nilotinib.
- Author
-
Yamashita Y, Morita S, Hosoi H, Kobata H, Kishimoto S, Ishibashi T, Mishima H, Kinoshita A, Backes BJ, Yoshiura KI, Papa FR, Sonoki T, and Tamura S
- Subjects
- Adult, Aged, Apoptosis, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism, Cell Movement, Cell Proliferation, Cross-Sectional Studies, Female, Follow-Up Studies, Humans, Male, Middle Aged, Multiple Myeloma pathology, Prognosis, Pyrazines administration & dosage, Pyrimidines administration & dosage, Retrospective Studies, Tumor Cells, Cultured, Antineoplastic Combined Chemotherapy Protocols pharmacology, Endoribonucleases antagonists & inhibitors, Gene Expression Regulation, Neoplastic drug effects, Molecular Targeted Therapy, Multiple Myeloma drug therapy, Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Background: Inositol-requiring enzyme 1α (IRE1α), along with protein kinase R-like endoplasmic reticulum kinase (PERK), is a principal regulator of the unfolded protein response (UPR). Recently, the 'mono'-specific IRE1α inhibitor, kinase-inhibiting RNase attenuator 6 (KIRA6), demonstrated a promising effect against multiple myeloma (MM). Side-stepping the clinical translation, a detailed UPR phenotype in patients with MM and the mechanisms of how KIRA8 works in MM remains unclear., Methods: We characterized UPR phenotypes in the bone marrow of patients with newly diagnosed MM. Then, in human MM cells we analyzed the possible anti-tumor mechanisms of KIRA8 and a Food and Drug Administration (FDA)-approved drug, nilotinib, which we recently identified as having a strong inhibitory effect against IRE1α activity. Finally, we performed an RNA-sequence analysis to detect key IRE1α-related molecules against MM., Results: We illustrated the dominant induction of adaptive UPR markers under IRE1α over the PERK pathway in patients with MM. In human MM cells, KIRA8 decreased cell viability and induced apoptosis, along with the induction of C/EBP homologous protein (CHOP); its combination with bortezomib exhibited more anti-myeloma effects than KIRA8 alone. Nilotinib exerted a similar effect compared with KIRA8. RNA-sequencing identified Polo-like kinase 2 ( PLK2 ) as a KIRA8-suppressed gene. Specifically, the IRE1α overexpression induced PLK2 expression, which was decreased by KIRA8. KIRA8 and PLK2 inhibition exerted anti-myeloma effects with apoptosis induction and the regulation of cell proliferation. Finally, PLK2 was pathologically confirmed to be highly expressed in patients with MM., Conclusion: Dominant activation of adaptive IRE1α was established in patients with MM. Both KIRA8 and nilotinib exhibited anti-myeloma effects, which were enhanced by bortezomib. Adaptive IRE1α signaling and PLK2 could be potential therapeutic targets and biomarkers in MM.
- Published
- 2020
- Full Text
- View/download PDF
8. Development of a Chemical Toolset for Studying the Paralog-Specific Function of IRE1.
- Author
-
Feldman HC, Vidadala VN, Potter ZE, Papa FR, Backes BJ, and Maly DJ
- Subjects
- Allosteric Regulation, Animals, Endoplasmic Reticulum Stress, Endoribonucleases metabolism, Humans, Protein Serine-Threonine Kinases metabolism, Ribonucleases metabolism, Endoribonucleases physiology, Protein Serine-Threonine Kinases physiology
- Abstract
The dual kinase endoribonuclease IRE1 is a master regulator of cell fate decisions in cells experiencing endoplasmic reticulum (ER) stress. In mammalian cells, there are two paralogs of IRE1: IRE1α and IRE1β. While IRE1α has been extensively studied, much less is understood about IRE1β and its role in signaling. In addition, whether the regulation of IRE1β's enzymatic activities varies compared to IRE1α is not known. Here, we show that the RNase domain of IRE1β is enzymatically active and capable of cleaving an XBP1 RNA mini-substrate in vitro . Using ATP-competitive inhibitors, we find that, like IRE1α, there is an allosteric relationship between the kinase and RNase domains of IRE1β. This allowed us to develop a novel toolset of both paralog specific and dual-IRE1α/β kinase inhibitors that attenuate RNase activity (KIRAs). Using sequence alignments of IRE1α and IRE1β, we propose a model for paralog-selective inhibition through interactions with nonconserved residues that differentiate the ATP-binding pockets of IRE1α and IRE1β.
- Published
- 2019
- Full Text
- View/download PDF
9. Parallel Signaling through IRE1α and PERK Regulates Pancreatic Neuroendocrine Tumor Growth and Survival.
- Author
-
Moore PC, Qi JY, Thamsen M, Ghosh R, Peng J, Gliedt MJ, Meza-Acevedo R, Warren RE, Hiniker A, Kim GE, Maly DJ, Backes BJ, Papa FR, and Oakes SA
- Subjects
- Adenine analogs & derivatives, Adenine pharmacology, Adenine therapeutic use, Animals, Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Disease Models, Animal, Endoplasmic Reticulum Stress drug effects, Endoribonucleases metabolism, Female, Humans, Indoles pharmacology, Indoles therapeutic use, Mice, Mice, Transgenic, Neuroendocrine Tumors genetics, Neuroendocrine Tumors pathology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Protein Kinase Inhibitors therapeutic use, Protein Serine-Threonine Kinases metabolism, Signal Transduction drug effects, Unfolded Protein Response drug effects, Xenograft Model Antitumor Assays, eIF-2 Kinase metabolism, Endoribonucleases antagonists & inhibitors, Neuroendocrine Tumors drug therapy, Pancreatic Neoplasms drug therapy, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors, eIF-2 Kinase antagonists & inhibitors
- Abstract
Master regulators of the unfolded protein response (UPR), IRE1α and PERK, promote adaptation or apoptosis depending on the level of endoplasmic reticulum (ER) stress. Although the UPR is activated in many cancers, its effects on tumor growth remain unclear. Derived from endocrine cells, pancreatic neuroendocrine tumors (PanNET) universally hypersecrete one or more peptide hormones, likely sensitizing these cells to high ER protein-folding stress. To assess whether targeting the UPR is a viable therapeutic strategy, we analyzed human PanNET samples and found evidence of elevated ER stress and UPR activation. Genetic and pharmacologic modulation of IRE1α and PERK in cultured cells, xenograft, and spontaneous genetic (RIP-Tag2) mouse models of PanNETs revealed that UPR signaling was optimized for adaptation and that inhibiting either IRE1α or PERK led to hyperactivation and apoptotic signaling through the reciprocal arm, thereby halting tumor growth and survival. These results provide a strong rationale for therapeutically targeting the UPR in PanNETs and other cancers with elevated ER stress. SIGNIFICANCE: The UPR is upregulated in pancreatic neuroendocrine tumors and its inhibition significantly reduces tumor growth in preclinical models, providing strong rationale for targeting the UPR in these cancers., (©2019 American Association for Cancer Research.)
- Published
- 2019
- Full Text
- View/download PDF
10. Small molecule inhibition of IRE1α kinase/RNase has anti-fibrotic effects in the lung.
- Author
-
Thamsen M, Ghosh R, Auyeung VC, Brumwell A, Chapman HA, Backes BJ, Perara G, Maly DJ, Sheppard D, and Papa FR
- Subjects
- Alveolar Epithelial Cells metabolism, Alveolar Epithelial Cells pathology, Animals, Apoptosis drug effects, Cell Line, Endoplasmic Reticulum Stress drug effects, Fibrosis metabolism, Fibrosis pathology, Lung metabolism, Lung pathology, Mice, Protein Kinase Inhibitors therapeutic use, Unfolded Protein Response drug effects, Alveolar Epithelial Cells drug effects, Endoribonucleases antagonists & inhibitors, Fibrosis drug therapy, Lung drug effects, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Endoplasmic reticulum stress (ER stress) has been implicated in the pathogenesis of idiopathic pulmonary fibrosis (IPF), a disease of progressive fibrosis and respiratory failure. ER stress activates a signaling pathway called the unfolded protein response (UPR) that either restores homeostasis or promotes apoptosis. The bifunctional kinase/RNase IRE1α is a UPR sensor/effector that promotes apoptosis if ER stress remains high and irremediable (i.e., a "terminal" UPR). Using multiple small molecule inhibitors against IRE1α, we show that ER stress-induced apoptosis of murine alveolar epithelial cells can be mitigated in vitro. In vivo, we show that bleomycin exposure to murine lungs causes early ER stress to activate IRE1α and the terminal UPR prior to development of pulmonary fibrosis. Small-molecule IRE1α kinase-inhibiting RNase attenuators (KIRAs) that we developed were used to evaluate the contribution of IRE1α activation to bleomycin-induced pulmonary fibrosis. One such KIRA-KIRA7-provided systemically to mice at the time of bleomycin exposure decreases terminal UPR signaling and prevents lung fibrosis. Administration of KIRA7 14 days after bleomycin exposure even promoted the reversal of established fibrosis. Finally, we show that KIRA8, a nanomolar-potent, monoselective KIRA compound derived from a completely different scaffold than KIRA7, likewise promoted reversal of established fibrosis. These results demonstrate that IRE1α may be a promising target in pulmonary fibrosis and that kinase inhibitors of IRE1α may eventually be developed into efficacious anti-fibrotic drugs., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
11. Fibroblast-specific inhibition of TGF-β1 signaling attenuates lung and tumor fibrosis.
- Author
-
Wei Y, Kim TJ, Peng DH, Duan D, Gibbons DL, Yamauchi M, Jackson JR, Le Saux CJ, Calhoun C, Peters J, Derynck R, Backes BJ, and Chapman HA
- Subjects
- A549 Cells, Amino Acid Oxidoreductases genetics, Amino Acid Oxidoreductases metabolism, Animals, Drug Screening Assays, Antitumor, Fibroblasts pathology, Humans, Mice, Neoplasm Metastasis, Neoplasm Proteins genetics, Phenols chemistry, Phenols pharmacology, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Receptor, Transforming Growth Factor-beta Type I, Receptors, Transforming Growth Factor beta genetics, Receptors, Transforming Growth Factor beta metabolism, Transforming Growth Factor beta1 antagonists & inhibitors, Transforming Growth Factor beta1 genetics, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Epithelial-Mesenchymal Transition, Fibroblasts metabolism, Lung Neoplasms drug therapy, Lung Neoplasms genetics, Lung Neoplasms metabolism, Neoplasm Proteins antagonists & inhibitors, Neoplasm Proteins metabolism, Protein Serine-Threonine Kinases antagonists & inhibitors, Pulmonary Fibrosis drug therapy, Pulmonary Fibrosis genetics, Pulmonary Fibrosis metabolism, Pulmonary Fibrosis pathology, Receptors, Transforming Growth Factor beta antagonists & inhibitors, Signal Transduction drug effects, Transforming Growth Factor beta1 metabolism
- Abstract
TGF-β1 signaling is a critical driver of collagen accumulation and fibrotic disease but also a vital suppressor of inflammation and epithelial cell proliferation. The nature of this multifunctional cytokine has limited the development of global TGF-β1 signaling inhibitors as therapeutic agents. We conducted phenotypic screens for small molecules that inhibit TGF-β1-induced epithelial-mesenchymal transition without immediate TGF-β1 receptor (TβR) kinase inhibition. We identified trihydroxyphenolic compounds as potent blockers of TGF-β1 responses (IC50 ~50 nM), Snail1 expression, and collagen deposition in vivo in models of pulmonary fibrosis and collagen-dependent lung cancer metastasis. Remarkably, the functional effects of trihydroxyphenolics required the presence of active lysyl oxidase-like 2 (LOXL2), thereby limiting effects to fibroblasts or cancer cells, the major LOXL2 producers. Mechanistic studies revealed that trihydroxyphenolics induce auto-oxidation of a LOXL2/3-specific lysine (K731) in a time-dependent reaction that irreversibly inhibits LOXL2 and converts the trihydrophenolic to a previously undescribed metabolite that directly inhibits TβRI kinase. Combined inhibition of LOXL2 and TβRI activities by trihydrophenolics resulted in potent blockade of pathological collagen accumulation in vivo without the toxicities associated with global inhibitors. These findings elucidate a therapeutic approach to attenuate fibrosis and the disease-promoting effects of tissue stiffness by specifically targeting TβRI kinase in LOXL2-expressing cells.
- Published
- 2017
- Full Text
- View/download PDF
12. Targeting ABL-IRE1α Signaling Spares ER-Stressed Pancreatic β Cells to Reverse Autoimmune Diabetes.
- Author
-
Morita S, Villalta SA, Feldman HC, Register AC, Rosenthal W, Hoffmann-Petersen IT, Mehdizadeh M, Ghosh R, Wang L, Colon-Negron K, Meza-Acevedo R, Backes BJ, Maly DJ, Bluestone JA, and Papa FR
- Published
- 2017
- Full Text
- View/download PDF
13. Structural and Functional Analysis of the Allosteric Inhibition of IRE1α with ATP-Competitive Ligands.
- Author
-
Feldman HC, Tong M, Wang L, Meza-Acevedo R, Gobillot TA, Lebedev I, Gliedt MJ, Hari SB, Mitra AK, Backes BJ, Papa FR, Seeliger MA, and Maly DJ
- Subjects
- Allosteric Regulation, Binding, Competitive, Endoplasmic Reticulum Stress, Endoribonucleases antagonists & inhibitors, Endoribonucleases chemistry, Humans, Ligands, Molecular Structure, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases chemistry, Ribonucleases antagonists & inhibitors, Structure-Activity Relationship, Adenosine Triphosphate metabolism, Endoribonucleases metabolism, Protein Serine-Threonine Kinases metabolism
- Abstract
The accumulation of unfolded proteins under endoplasmic reticulum (ER) stress leads to the activation of the multidomain protein sensor IRE1α as part of the unfolded protein response (UPR). Clustering of IRE1α lumenal domains in the presence of unfolded proteins promotes kinase trans-autophosphorylation in the cytosol and subsequent RNase domain activation. Interestingly, there is an allosteric relationship between the kinase and RNase domains of IRE1α, which allows ATP-competitive inhibitors to modulate the activity of the RNase domain. Here, we use kinase inhibitors to study how ATP-binding site conformation affects the activity of the RNase domain of IRE1α. We find that diverse ATP-competitive inhibitors of IRE1α promote dimerization and activation of RNase activity despite blocking kinase autophosphorylation. In contrast, a subset of ATP-competitive ligands, which we call KIRAs, allosterically inactivate the RNase domain through the kinase domain by stabilizing monomeric IRE1α. Further insight into how ATP-competitive inhibitors are able to divergently modulate the RNase domain through the kinase domain was gained by obtaining the first structure of apo human IRE1α in the RNase active back-to-back dimer conformation. Comparison of this structure with other existing structures of IRE1α and integration of our extensive structure activity relationship (SAR) data has led us to formulate a model to rationalize how ATP-binding site ligands are able to control the IRE1α oligomeric state and subsequent RNase domain activity., Competing Interests: Bradley J. Backes, Feroz R. Papa, and Dustin J. Maly are scientific co-founders, equity holders, and paid consultants for OptiKira L. L. C.
- Published
- 2016
- Full Text
- View/download PDF
14. Allosteric inhibition of the IRE1α RNase preserves cell viability and function during endoplasmic reticulum stress.
- Author
-
Ghosh R, Wang L, Wang ES, Perera BG, Igbaria A, Morita S, Prado K, Thamsen M, Caswell D, Macias H, Weiberth KF, Gliedt MJ, Alavi MV, Hari SB, Mitra AK, Bhhatarai B, Schürer SC, Snapp EL, Gould DB, German MS, Backes BJ, Maly DJ, Oakes SA, and Papa FR
- Subjects
- Allosteric Regulation, Animals, Apoptosis drug effects, Cell Line, Endoribonucleases chemistry, Endoribonucleases metabolism, Enzyme Activation drug effects, Humans, Islets of Langerhans metabolism, Male, Mice, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases metabolism, Rats, Retina metabolism, Ribonucleases antagonists & inhibitors, Endoplasmic Reticulum Stress, Endoribonucleases antagonists & inhibitors, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Depending on endoplasmic reticulum (ER) stress levels, the ER transmembrane multidomain protein IRE1α promotes either adaptation or apoptosis. Unfolded ER proteins cause IRE1α lumenal domain homo-oligomerization, inducing trans autophosphorylation that further drives homo-oligomerization of its cytosolic kinase/endoribonuclease (RNase) domains to activate mRNA splicing of adaptive XBP1 transcription factor. However, under high/chronic ER stress, IRE1α surpasses an oligomerization threshold that expands RNase substrate repertoire to many ER-localized mRNAs, leading to apoptosis. To modulate these effects, we developed ATP-competitive IRE1α Kinase-Inhibiting RNase Attenuators-KIRAs-that allosterically inhibit IRE1α's RNase by breaking oligomers. One optimized KIRA, KIRA6, inhibits IRE1α in vivo and promotes cell survival under ER stress. Intravitreally, KIRA6 preserves photoreceptor functional viability in rat models of ER stress-induced retinal degeneration. Systemically, KIRA6 preserves pancreatic β cells, increases insulin, and reduces hyperglycemia in Akita diabetic mice. Thus, IRE1α powerfully controls cell fate but can itself be controlled with small molecules to reduce cell degeneration., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
15. Divergent allosteric control of the IRE1α endoribonuclease using kinase inhibitors.
- Author
-
Wang L, Perera BG, Hari SB, Bhhatarai B, Backes BJ, Seeliger MA, Schürer SC, Oakes SA, Papa FR, and Maly DJ
- Subjects
- Adaptor Proteins, Signal Transducing, Catalysis, Cells, Cultured, Cross-Linking Reagents, DNA-Binding Proteins metabolism, Down-Regulation drug effects, Endoplasmic Reticulum Stress physiology, Humans, Intracellular Signaling Peptides and Proteins, Isoenzymes antagonists & inhibitors, Isoenzymes metabolism, Molecular Conformation, Mutation genetics, Mutation physiology, Phosphorylation, RNA Splicing drug effects, Regulatory Factor X Transcription Factors, Ribonucleases metabolism, Transcription Factors metabolism, Unfolded Protein Response drug effects, Up-Regulation drug effects, X-Box Binding Protein 1, Endoribonucleases antagonists & inhibitors, Protein Kinase Inhibitors pharmacology, Protein Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Under endoplasmic reticulum stress, unfolded protein accumulation leads to activation of the endoplasmic reticulum transmembrane kinase/endoRNase (RNase) IRE1α. IRE1α oligomerizes, autophosphorylates and initiates splicing of XBP1 mRNA, thus triggering the unfolded protein response (UPR). Here we show that IRE1α's kinase-controlled RNase can be regulated in two distinct modes with kinase inhibitors: one class of ligands occupies IRE1α's kinase ATP-binding site to activate RNase-mediated XBP1 mRNA splicing even without upstream endoplasmic reticulum stress, whereas a second class can inhibit the RNase through the same ATP-binding site, even under endoplasmic reticulum stress. Thus, alternative kinase conformations stabilized by distinct classes of ATP-competitive inhibitors can cause allosteric switching of IRE1α's RNase--either on or off. As dysregulation of the UPR has been implicated in a variety of cell degenerative and neoplastic disorders, small-molecule control over IRE1α should advance efforts to understand the UPR's role in pathophysiology and to develop drugs for endoplasmic reticulum stress-related diseases.
- Published
- 2012
- Full Text
- View/download PDF
16. IRE1α induces thioredoxin-interacting protein to activate the NLRP3 inflammasome and promote programmed cell death under irremediable ER stress.
- Author
-
Lerner AG, Upton JP, Praveen PV, Ghosh R, Nakagawa Y, Igbaria A, Shen S, Nguyen V, Backes BJ, Heiman M, Heintz N, Greengard P, Hui S, Tang Q, Trusina A, Oakes SA, and Papa FR
- Subjects
- Animals, Blotting, Western, Cell Line, DNA Primers genetics, Flow Cytometry, Humans, Interleukin-1beta metabolism, Mice, Mice, Inbred C57BL, NLR Family, Pyrin Domain-Containing 3 Protein, Real-Time Polymerase Chain Reaction, Apoptosis physiology, Carrier Proteins metabolism, Endoplasmic Reticulum Stress physiology, Endoribonucleases metabolism, Inflammasomes metabolism, Protein Serine-Threonine Kinases metabolism, Thioredoxins metabolism, Unfolded Protein Response physiology
- Abstract
When unfolded proteins accumulate to irremediably high levels within the endoplasmic reticulum (ER), intracellular signaling pathways called the unfolded protein response (UPR) become hyperactivated to cause programmed cell death. We discovered that thioredoxin-interacting protein (TXNIP) is a critical node in this "terminal UPR." TXNIP becomes rapidly induced by IRE1α, an ER bifunctional kinase/endoribonuclease (RNase). Hyperactivated IRE1α increases TXNIP mRNA stability by reducing levels of a TXNIP destabilizing microRNA, miR-17. In turn, elevated TXNIP protein activates the NLRP3 inflammasome, causing procaspase-1 cleavage and interleukin 1β (IL-1β) secretion. Txnip gene deletion reduces pancreatic β cell death during ER stress and suppresses diabetes caused by proinsulin misfolding in the Akita mouse. Finally, small molecule IRE1α RNase inhibitors suppress TXNIP production to block IL-1β secretion. In summary, the IRE1α-TXNIP pathway is used in the terminal UPR to promote sterile inflammation and programmed cell death and may be targeted to develop effective treatments for cell degenerative diseases., (Copyright © 2012 Elsevier Inc. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
17. IRE1alpha kinase activation modes control alternate endoribonuclease outputs to determine divergent cell fates.
- Author
-
Han D, Lerner AG, Vande Walle L, Upton JP, Xu W, Hagen A, Backes BJ, Oakes SA, and Papa FR
- Subjects
- Animals, Cell Line, Cell Line, Tumor, Cells metabolism, Endoplasmic Reticulum metabolism, Insulin genetics, Multienzyme Complexes, Protein Folding, Protein Serine-Threonine Kinases, RNA Stability, Rats, Ribonucleases, Endoribonucleases metabolism
- Abstract
During endoplasmic reticulum (ER) stress, homeostatic signaling through the unfolded protein response (UPR) augments ER protein-folding capacity. If homeostasis is not restored, the UPR triggers apoptosis. We found that the ER transmembrane kinase/endoribonuclease (RNase) IRE1alpha is a key component of this apoptotic switch. ER stress induces IRE1alpha kinase autophosphorylation, activating the RNase to splice XBP1 mRNA and produce the homeostatic transcription factor XBP1s. Under ER stress--or forced autophosphorylation--IRE1alpha's RNase also causes endonucleolytic decay of many ER-localized mRNAs, including those encoding chaperones, as early events culminating in apoptosis. Using chemical genetics, we show that kinase inhibitors bypass autophosphorylation to activate the RNase by an alternate mode that enforces XBP1 splicing and averts mRNA decay and apoptosis. Alternate RNase activation by kinase-inhibited IRE1alpha can be reconstituted in vitro. We propose that divergent cell fates during ER stress hinge on a balance between IRE1alpha RNase outputs that can be tilted with kinase inhibitors to favor survival.
- Published
- 2009
- Full Text
- View/download PDF
18. Effect of bradykinin metabolism inhibitors on evoked hypotension in rats: rank efficacy of enzymes associated with bradykinin-mediated angioedema.
- Author
-
Fryer RM, Segreti J, Banfor PN, Widomski DL, Backes BJ, Lin CW, Ballaron SJ, Cox BF, Trevillyan JM, Reinhart GA, and von Geldern TW
- Subjects
- Aminopeptidases antagonists & inhibitors, Angiotensin-Converting Enzyme Inhibitors administration & dosage, Angiotensin-Converting Enzyme Inhibitors pharmacology, Animals, Bradykinin drug effects, Dose-Response Relationship, Drug, Drug Evaluation, Preclinical methods, Indans pharmacology, Lisinopril pharmacology, Male, Neprilysin antagonists & inhibitors, Peptides pharmacology, Propionates pharmacology, Pyridines administration & dosage, Pyridines pharmacology, Rats, Rats, Sprague-Dawley, Thiazepines administration & dosage, Thiazepines pharmacology, Angioedema etiology, Bradykinin metabolism, Enzyme Inhibitors pharmacology, Hypotension etiology
- Abstract
Background and Purpose: Inhibition of bradykinin metabolizing enzymes (BMEs) can cause acute angioedema, as demonstrated in a recent clinical trial in patients administered the antihypertensive, omapatrilat. However, the relative contribution of specific BMEs to this effect is unclear and confounded by the lack of a predictive pre-clinical model of angioedema., Experimental Approach: Rats were instrumented to record blood pressure and heart rate; inhibitors were infused for 35 min and bradykinin was infused during the last 5 min to elicit hypotension, as a functional marker of circulating bradykinin and relative angioedema risk., Key Results: In the presence of omapatrilat bradykinin produced dose-dependent hypotension, an effect abolished by B(2) blockade. In the presence of lisinopril (ACE inhibitor), but not candoxatril (NEP inhibitor) or apstatin (APP inhibitor), bradykinin also elicited hypotension. Lisinopril-mediated hypotension was unchanged with concomitant blockade of NEP or NEP/DPPIV (candoxatril+A-899301). However, hypotension was enhanced upon concomitant blockade of APP and further intensified in the presence of NEP inhibition to values not different from omapatrilat alone., Conclusions and Implications: We demonstrated that bradykinin is degraded in vivo with an enzyme rank-efficacy of ACE>APP>>NEP or DPPIV. These results suggest the effects of omapatrilat are mediated by inhibition of three BMEs, ACE/APP/NEP. However, dual inhibition of ACE/NEP or ACE/NEP/DPPIV elicits no increased risk of angioedema compared to ACE inhibition alone. Thus, novel BME inhibitors must display no activity against APP to avoid angioedema risk due to high prevalence of ACE inhibitor therapy in patients with diabetes and cardiovascular disease.
- Published
- 2008
- Full Text
- View/download PDF
19. Synthesis of C-11 modified mifepristone analog libraries.
- Author
-
Hamilton GL and Backes BJ
- Subjects
- Aniline Compounds chemistry, Hormone Antagonists chemical synthesis, Mifepristone chemistry, Models, Biological, Chemistry, Pharmaceutical methods, Mifepristone analogs & derivatives, Mifepristone chemical synthesis, Peptide Library
- Abstract
Substitution of the C-11 aniline of mifepristone can provide compounds with altered pharmacokinetic and pharmacodynamic (PK/PD) profiles that may find use for new indications. The development of new steroid intermediates and specialized library synthesis methods were required to enable the efficient preparation of structurally complex C-11 modified mifepristone analogs.
- Published
- 2007
- Full Text
- View/download PDF
20. Discovery and structure-activity relationships of piperidinone- and piperidine-constrained phenethylamines as novel, potent, and selective dipeptidyl peptidase IV inhibitors.
- Author
-
Pei Z, Li X, von Geldern TW, Longenecker K, Pireh D, Stewart KD, Backes BJ, Lai C, Lubben TH, Ballaron SJ, Beno DW, Kempf-Grote AJ, Sham HL, and Trevillyan JM
- Subjects
- Animals, Biological Availability, Crystallography, X-Ray, Dipeptidyl Peptidase 4, Humans, Molecular Conformation, Phenethylamines pharmacokinetics, Phenethylamines pharmacology, Piperidines pharmacokinetics, Piperidines pharmacology, Piperidones chemical synthesis, Piperidones pharmacokinetics, Piperidones pharmacology, Rats, Stereoisomerism, Structure-Activity Relationship, Adenosine Deaminase Inhibitors, Dipeptidyl-Peptidase IV Inhibitors, Glycoproteins antagonists & inhibitors, Phenethylamines chemical synthesis, Piperidines chemical synthesis
- Abstract
Dipeptidyl peptidase IV (DPP4) inhibitors are emerging as a new class of therapeutic agents for the treatment of type 2 diabetes. They exert their beneficial effects by increasing the levels of active glucagon-like peptide-1 and glucose-dependent insulinotropic peptide, which are two important incretins for glucose homeostasis. Starting from a high-throughput screening hit, we were able to identify a series of piperidinone- and piperidine-constrained phenethylamines as novel DPP4 inhibitors. Optimized compounds are potent, selective, and have good pharmacokinetic profiles.
- Published
- 2007
- Full Text
- View/download PDF
21. Pyrrolidine-constrained phenethylamines: The design of potent, selective, and pharmacologically efficacious dipeptidyl peptidase IV (DPP4) inhibitors from a lead-like screening hit.
- Author
-
Backes BJ, Longenecker K, Hamilton GL, Stewart K, Lai C, Kopecka H, von Geldern TW, Madar DJ, Pei Z, Lubben TH, Zinker BA, Tian Z, Ballaron SJ, Stashko MA, Mika AK, Beno DW, Kempf-Grote AJ, Black-Schaefer C, Sham HL, and Trevillyan JM
- Subjects
- Animals, Blood Glucose metabolism, Cyclohexenes chemistry, Diabetes Mellitus, Type 2 drug therapy, Drug Design, Enzyme Inhibitors chemistry, Female, Hypoglycemic Agents pharmacology, Models, Chemical, Molecular Conformation, Phenethylamines chemistry, Pyrrolidines chemistry, Rats, Chemistry, Pharmaceutical methods, Dipeptidyl-Peptidase IV Inhibitors, Enzyme Inhibitors chemical synthesis, Phenethylamines chemical synthesis, Pyrrolidines chemical synthesis
- Abstract
A novel series of pyrrolidine-constrained phenethylamines were developed as dipeptidyl peptidase IV (DPP4) inhibitors for the treatment of type 2 diabetes. The cyclohexene ring of lead-like screening hit 5 was replaced with a pyrrolidine to enable parallel chemistry, and protein co-crystal structural data guided the optimization of N-substituents. Employing this strategy, a >400x improvement in potency over the initial hit was realized in rapid fashion. Optimized compounds are potent and selective inhibitors with excellent pharmacokinetic profiles. Compound 30 was efficacious in vivo, lowering blood glucose in ZDF rats that were allowed to feed freely on a mixed meal.
- Published
- 2007
- Full Text
- View/download PDF
22. Parallel strategies for the preparation and selection of liver-targeted glucocorticoid receptor antagonists.
- Author
-
Backes BJ, Hamilton GL, Nguyen P, Wilcox D, Fung S, Wang J, Grynfarb M, Goos-Nilsson A, Jacobson PB, and von Geldern TW
- Subjects
- Animals, Glucocorticoids chemical synthesis, Rats, Rats, Inbred Strains, Glucocorticoids chemistry, Glucocorticoids pharmacokinetics, Liver metabolism, Mifepristone analogs & derivatives, Receptors, Glucocorticoid antagonists & inhibitors
- Abstract
Libraries of mifepristone analogs, MP-Acids, were designed and synthesized to increase the chances of identifying GR antagonists that possess liver-selective pharmacological profiles. MP-Acids were uniformly potent GR antagonists in binding and in cell-based functional assays. A high throughput pharmacokinetic selection strategy that employs the cassette dosing of MP-Acids was developed to identify liver-targeting compounds. Thus, resource-intensive in vivo assays to measure liver-selective pharmacology were enriched with GR antagonists that achieve high concentrations in the liver.
- Published
- 2007
- Full Text
- View/download PDF
23. Discovery of ((4R,5S)-5-amino-4-(2,4,5- trifluorophenyl)cyclohex-1-enyl)-(3- (trifluoromethyl)-5,6-dihydro- [1,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl)methanone (ABT-341), a highly potent, selective, orally efficacious, and safe dipeptidyl peptidase IV inhibitor for the treatment of type 2 diabetes.
- Author
-
Pei Z, Li X, von Geldern TW, Madar DJ, Longenecker K, Yong H, Lubben TH, Stewart KD, Zinker BA, Backes BJ, Judd AS, Mulhern M, Ballaron SJ, Stashko MA, Mika AK, Beno DW, Reinhart GA, Fryer RM, Preusser LC, Kempf-Grote AJ, Sham HL, and Trevillyan JM
- Subjects
- Animals, Biphenyl Compounds chemical synthesis, Biphenyl Compounds pharmacokinetics, Cyclohexenes chemistry, Diabetes Mellitus, Type 2 genetics, Dose-Response Relationship, Drug, Drug Design, Drug Evaluation, Preclinical, Female, Hypoglycemic Agents chemical synthesis, Hypoglycemic Agents pharmacokinetics, Models, Molecular, Rats, Rats, Zucker, Serine Proteinase Inhibitors chemical synthesis, Serine Proteinase Inhibitors pharmacokinetics, Structure-Activity Relationship, Triazoles chemical synthesis, Triazoles pharmacokinetics, X-Ray Diffraction, Biphenyl Compounds pharmacology, Diabetes Mellitus, Type 2 drug therapy, Dipeptidyl Peptidase 4 metabolism, Hypoglycemic Agents pharmacology, Serine Proteinase Inhibitors pharmacology, Triazoles pharmacology
- Abstract
Dipeptidyl peptidase IV (DPP4) deactivates glucose-regulating hormones such as GLP-1 and GIP, thus, DPP4 inhibition has become a useful therapy for type 2 diabetes. Optimization of the high-throughput screening lead 6 led to the discovery of 25 (ABT-341), a highly potent, selective, and orally bioavailable DPP4 inhibitor. When dosed orally, 25 dose-dependently reduced glucose excursion in ZDF rats. Amide 25 is safe in a battery of in vitro and in vivo tests and may represent a new therapeutic agent for the treatment of type 2 diabetes.
- Published
- 2006
- Full Text
- View/download PDF
24. A strategy to profile prime and non-prime proteolytic substrate specificity.
- Author
-
Petrassi HM, Williams JA, Li J, Tumanut C, Ek J, Nakai T, Masick B, Backes BJ, and Harris JL
- Subjects
- Amino Acid Sequence, Caspase 3, Computer Simulation, Coumarins chemistry, Fluorescent Dyes metabolism, Humans, Models, Molecular, Molecular Sequence Data, Substrate Specificity, Caspases metabolism, Combinatorial Chemistry Techniques, Coumarins metabolism, Oligopeptides chemical synthesis, Oligopeptides metabolism, Thrombin metabolism
- Abstract
A strategy was developed to determine the prime and non-prime substrate specificity of serine, threonine and cysteine proteases. ACC positional scanning technology was employed to determine the P4-P1 non-prime site substrate specificity. The data was used to synthesize biased donor-quencher positional scanning libraries to profile the P1'-P4' prime site substrate specificity. Directed sorting using the Irori Nanokan system allowed for the archiving of multiple P1'-P4' positional scanning libraries. From these libraries focused donor-quencher libraries incorporating P4-P1 data for each protease under study could be rapidly prepared. The profiling of thrombin and caspase-3 P4-P4' substrate specificity, comparison of the library specificity data to single substrates, and the analysis of physiological cleavage sites are described.
- Published
- 2005
- Full Text
- View/download PDF
25. Purifying the masses: integrating prepurification quality control, high-throughput LC/MS purification, and compound plating to feed high-throughput screening.
- Author
-
Isbell JJ, Zhou Y, Guintu C, Rynd M, Jiang S, Petrov D, Micklash K, Mainquist J, Ek J, Chang J, Weselak M, Backes BJ, Brailsford A, and Shave D
- Subjects
- Autoanalysis, Chromatography, High Pressure Liquid, Indicators and Reagents, Mass Spectrometry, Pharmaceutical Preparations standards, Quality Control, Robotics, Combinatorial Chemistry Techniques methods, Pharmaceutical Preparations isolation & purification
- Abstract
In this paper we report using a parallel, four-channel HPLC/MUX/MS purification system, the Purification Factory, to purify thousands of compounds destined for high-throughput screening in a single month. The maximum sample throughput during this 20-workday month was 704 samples/day. Since this purification throughput exceeded the postpurification sample and data handling capabilities provided by commercial solutions, a custom-integrated solution was designed to address these shortcomings. In this paper we detail the key improvements in automation, solvent handling, and sample handling logistics implemented to sustain a mean throughput of 528 samples/day over a multimonth time period.
- Published
- 2005
- Full Text
- View/download PDF
26. Biochemical characterization of prostasin, a channel activating protease.
- Author
-
Shipway A, Danahay H, Williams JA, Tully DC, Backes BJ, and Harris JL
- Subjects
- Animals, Anions, Arginine chemistry, Binding Sites, Blotting, Western, Cations, Cell Line, Cloning, Molecular, DNA, Complementary metabolism, Electrophoresis, Polyacrylamide Gel, Epithelial Sodium Channels, Gene Library, Histidine chemistry, Humans, Hydrogen-Ion Concentration, Insecta, Ions, Kinetics, Lysine chemistry, Metals chemistry, Models, Genetic, Peptide Library, Protein Binding, Protein Structure, Tertiary, Recombinant Proteins chemistry, Sodium Channels chemistry, Substrate Specificity, Zinc chemistry, Peptide Hydrolases metabolism, Serine Endopeptidases chemistry, Serine Endopeptidases physiology
- Abstract
Human prostasin was recently identified as a potential regulator of epithelial sodium channel (ENaC) function. Through the use of positional scanning combinatorial substrate libraries, prostasin was shown to have a preference for poly-basic substrates: in position P4 preference was for arginine or lysine; in P3 preference was for histidine, lysine or arginine; in P2 preference was for basic or large hydrophobic amino acids; and in P1 preference was for arginine and lysine. P1', P2', and P3' displayed broad selectivity with the exception of a lack of activity for isoleucine, and P4' had a preference for small, unbranched, amino acids such as alanine and serine. A prostasin-preferred poly-basic cleavage site was found in the extracellular domains of the ENaC alpha- and beta-subunits, and may present a mechanism for prostasin activation. The absence of activity seen with substrates containing isoleucine in position P1' explains the inability of prostasin to autoactivate and suggests that prostasin proteolytic activity is regulated by an upstream protease. Prostasin activity was highly influenced by mono- and divalent metal ions which were potent inhibitors and substrate specific modulators of enzymatic activity. In the presence of sub-inhibitory concentrations of zinc, the activity of prostasin increased several-fold and its substrate specificity was significantly altered in favor of a strong preference for histidine in positions P3 or P4 of the substrate.
- Published
- 2004
- Full Text
- View/download PDF
27. Activity profile of dust mite allergen extract using substrate libraries and functional proteomic microarrays.
- Author
-
Harris J, Mason DE, Li J, Burdick KW, Backes BJ, Chen T, Shipway A, Van Heeke G, Gough L, Ghaemmaghami A, Shakib F, Debaene F, and Winssinger N
- Subjects
- Amino Acid Sequence, Antigens, Dermatophagoides isolation & purification, Antigens, Dermatophagoides metabolism, Cysteine Endopeptidases chemistry, Cysteine Endopeptidases genetics, Cysteine Endopeptidases metabolism, Enzyme Activation, Humans, Molecular Sequence Data, Protein Interaction Mapping methods, Protein Serine-Threonine Kinases chemistry, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Proteomics methods, Serine Endopeptidases chemistry, Serine Endopeptidases genetics, Serine Endopeptidases metabolism, Substrate Specificity genetics, Antigens, Dermatophagoides genetics, Oligonucleotide Array Sequence Analysis methods, Peptide Library
- Abstract
Enzymatic activity in the fecal droppings from the house dust mite has been postulated to contribute to the elicited allergic response. Screening dust mite extracts through 137,180 tetrapeptide fluorogenic substrates allowed for the characterization of proteolytic substrate specificity from the potential cysteine and serine proteases in the extract. The extract was further screened against a 4000 member peptide nucleic acid (PNA) encoded inhibitor library designed to target cysteine proteases using microarray detection. Affinity chromatography coupled with mass spectrometry identified Der p 1 as one of the proteases targeted by the PNA inhibitors in the dust mite lysate. A phenotypic readout of Der p 1 function in allergy progression was demonstrated by the inhibition of CD25 cleavage from T cells by dust mite extract that had been treated with the Der p 1 inhibitor identified from the PNA-encoded inhibitor library.
- Published
- 2004
- Full Text
- View/download PDF
28. Biochemical analysis of the 20 S proteasome of Trypanosoma brucei.
- Author
-
Wang CC, Bozdech Z, Liu CL, Shipway A, Backes BJ, Harris JL, and Bogyo M
- Subjects
- Amino Acid Sequence, Animals, Binding Sites, Catalytic Domain, Cysteine Endopeptidases metabolism, Molecular Sequence Data, Multienzyme Complexes metabolism, Proteasome Endopeptidase Complex, Protein Subunits, Substrate Specificity, Cysteine Endopeptidases chemistry, Multienzyme Complexes chemistry, Protozoan Proteins chemistry, Trypanosoma brucei brucei enzymology
- Abstract
We describe here biochemical characterization of the 20 S proteasome from the parasitic protozoan Trypanosoma brucei. Similar to the mammalian proteasome, the T. brucei proteasome is made up of seven alpha- and seven beta-subunits. Of the seven beta-type subunits, five contain pro-sequences that are proteolytically removed during assembly, and three of them are predicted to be catalytic based on primary sequence. Affinity labeling studies revealed that, unlike the mammalian proteasome where three beta-subunits were labeled by the affinity reagents, only two beta-subunits of the T. brucei proteasome were labeled in the complex. These two subunits corresponded to beta2 and beta5 subunits responsible for the trypsin-like and chymotrypsin-like proteolytic activities, respectively. Screening of a library of 137,180 tetrapeptide fluorogenic substrates against the T. brucei 20 S proteasome confirmed the nominal beta1-subunit (caspase-like or PGPH) activity and identified an overall substrate preference for hydrophobic residues at the P1 to P4 positions in a substrate. This overall stringency is relaxed in the 11 S regulator (PA26)-20 S proteasome complex, which shows both appreciable activities for cleavage after acidic amino acids and a broadened activity for cleavage after basic amino acids. The 20 S proteasome from T. brucei also shows appreciable activity for cleavage after P1-Gln that is minimally observed in the human counterpart. These results demonstrate the importance of substrate sequence specificity of the T. brucei proteasome and highlight its biochemical divergence from the human enzyme.
- Published
- 2003
- Full Text
- View/download PDF
29. Expedient solid-phase synthesis of fluorogenic protease substrates using the 7-amino-4-carbamoylmethylcoumarin (ACC) fluorophore.
- Author
-
Maly DJ, Leonetti F, Backes BJ, Dauber DS, Harris JL, Craik CS, and Ellman JA
- Subjects
- Amino Acids chemistry, Coumarins chemistry, Fluorescent Dyes chemistry, Magnetic Resonance Spectroscopy, Mass Spectrometry, Substrate Specificity, Coumarins metabolism, Fibrinolysin metabolism, Fluorescent Dyes metabolism, HIV Protease metabolism
- Abstract
A highly efficient solid-phase synthesis method for the preparation of fluorogenic protease substrates based upon the bifunctional leaving group 7-amino-4-carbamoylmethylcoumarin (ACC) is reported. Methods for the large-scale preparation of the novel fluorogenic leaving-group ACC are provided (Scheme 1). Detailed procedures are also provided for loading a diverse set of amino acids to support-bound ACC in good yields and with minimal racemization. Finally, procedures are included for the preparative synthesis of optimized ACC substrates for HIV-1 protease and plasmin.
- Published
- 2002
- Full Text
- View/download PDF
30. Substrate specificity of the human proteasome.
- Author
-
Harris JL, Alper PB, Li J, Rechsteiner M, and Backes BJ
- Subjects
- Amino Acid Sequence, Enzyme Activation, Histocompatibility Antigens Class I metabolism, Humans, Ligands, Molecular Sequence Data, Peptide Library, Proteasome Endopeptidase Complex, Structure-Activity Relationship, Substrate Specificity, Cysteine Endopeptidases metabolism, Multienzyme Complexes metabolism
- Abstract
Background: Regulated proteolysis by the proteasome is crucial for a broad array of cellular processes, from control of the cell cycle to production of antigens., Results: The rules governing the N-terminal primary and extended substrate specificity of the human 20S proteasome in the presence or absence of 11S proteasome activators (REGalpha/beta and REGgamma) have been elaborated using activity-based proteomic library tools., Conclusions: The 11S proteasome activators are shown to be important for both increasing the activity of the 20S proteasome and for altering its cleavage pattern and substrate specificity. These data also establish that the extended substrate specificity is an important factor for proteasomal cleavage. The specificities observed have features in common with major histocompatibility complex (MHC) class I ligands and can be used to improve the prediction of MHC class I restricted cytotoxic T-cell responses.
- Published
- 2001
- Full Text
- View/download PDF
31. Definition of the extended substrate specificity determinants for beta-tryptases I and II.
- Author
-
Harris JL, Niles A, Burdick K, Maffitt M, Backes BJ, Ellman JA, Kuntz I, Haak-Frendscho M, and Craik CS
- Subjects
- Humans, Pichia genetics, Recombinant Proteins metabolism, Serine Endopeptidases chemistry, Serine Proteinase Inhibitors pharmacology, Substrate Specificity, Tryptases, Isoenzymes metabolism, Serine Endopeptidases metabolism
- Abstract
Tryptases betaI and betaII were heterologously expressed and purified in yeast to functionally characterize the substrate specificity of each enzyme. Three positional scanning combinatorial tetrapeptide substrate libraries were used to determine the primary and extended substrate specificity of the proteases. Both enzymes have a strict primary preference for cleavage after the basic amino acids, lysine and arginine, with only a slight preference for lysine over arginine. betaI and betaII tryptase share similar extended substrate specificity, with preference for proline at P4, preference for arginine or lysine at P3, and P2 showing a slight preference for asparagine. Measurement of kinetic constants with multiple substrates designed for beta-tryptases reveal that selectivity is highly dependent on ground state substrate binding. Coupled with the functional determinants, structural determinants of tryptase substrate specificity were identified. Molecular docking of the preferred substrate sequence to the three-dimensional tetrameric tryptase structure reveals a novel extended substrate binding mode that involves interactions from two adjacent protomers, including P4 Thr-96', P3 Asp-60B' and Glu-217, and P1 Asp-189. Based on the determined substrate information, a mechanism-based tetrapeptide-chloromethylketone inhibitor was designed and shown to be a potent tryptase inhibitor. Finally, the cleavage sites of several physiologically relevant substrates of beta-tryptases show consistency with the specificity data presented here.
- Published
- 2001
- Full Text
- View/download PDF
32. From Split-Pool Libraries to Spatially Addressable Microarrays and Its Application to Functional Proteomic Profiling.
- Author
-
Winssinger N, Harris JL, Backes BJ, and Schultz PG
- Abstract
Small molecules encoded with peptidonucleic acid (PNA) were used to probe protein function in a microarray format. The PNA tag served to encode the synthetic history of the small molecule and to positionally encode the identity of the small molecule by its location upon hybridization to an oligonucleotide microarray., (© 2001 WILEY-VCH Verlag GmbH, Weinheim, Fed. Rep. of Germany.)
- Published
- 2001
- Full Text
- View/download PDF
33. Rapid and general profiling of protease specificity by using combinatorial fluorogenic substrate libraries.
- Author
-
Harris JL, Backes BJ, Leonetti F, Mahrus S, Ellman JA, and Craik CS
- Subjects
- Coumarins chemistry, Cysteine Endopeptidases metabolism, Serine Endopeptidases metabolism, Substrate Specificity, Combinatorial Chemistry Techniques, Coumarins metabolism, Endopeptidases metabolism, Fluorescent Dyes metabolism, Peptides chemical synthesis, Peptides metabolism
- Abstract
A method is presented for the preparation and use of fluorogenic peptide substrates that allows for the configuration of general substrate libraries to rapidly identify the primary and extended specificity of proteases. The substrates contain the fluorogenic leaving group 7-amino-4-carbamoylmethylcoumarin (ACC). Substrates incorporating the ACC leaving group show kinetic profiles comparable to those with the traditionally used 7-amino-4-methylcoumarin (AMC) leaving group. The bifunctional nature of ACC allows for the efficient production of single substrates and substrate libraries by using 9-fluorenylmethoxycarbonyl (Fmoc)-based solid-phase synthesis techniques. The approximately 3-fold-increased quantum yield of ACC over AMC permits reduction in enzyme and substrate concentrations. As a consequence, a greater number of substrates can be tolerated in a single assay, thus enabling an increase in the diversity space of the library. Soluble positional protease substrate libraries of 137, 180 and 6,859 members, possessing amino acid diversity at the P4-P3-P2-P1 and P4-P3-P2 positions, respectively, were constructed. Employing this screening method, we profiled the substrate specificities of a diverse array of proteases, including the serine proteases thrombin, plasmin, factor Xa, urokinase-type plasminogen activator, tissue plasminogen activator, granzyme B, trypsin, chymotrypsin, human neutrophil elastase, and the cysteine proteases papain and cruzain. The resulting profiles create a pharmacophoric portrayal of the proteases to aid in the design of selective substrates and potent inhibitors.
- Published
- 2000
- Full Text
- View/download PDF
34. Synthesis of positional-scanning libraries of fluorogenic peptide substrates to define the extended substrate specificity of plasmin and thrombin.
- Author
-
Backes BJ, Harris JL, Leonetti F, Craik CS, and Ellman JA
- Subjects
- Amino Acid Sequence, Computer Simulation, Fluorescent Dyes, Models, Molecular, Substrate Specificity, Combinatorial Chemistry Techniques, Fibrinolysin metabolism, Oligopeptides chemical synthesis, Oligopeptides metabolism, Thrombin metabolism
- Abstract
We have developed a strategy for the synthesis of positional-scanning synthetic combinatorial libraries (PS-SCL) that does not depend on the identity of the P1 substituent. To demonstrate the strategy, we synthesized a tetrapeptide positional library in which the P1 amino acid is held constant as a lysine and the P4-P3-P2 positions are positionally randomized. The 6,859 members of the library were synthesized on solid support with an alkane sulfonamide linker, and then displaced from the solid support by condensation with a fluorogenic 7-amino-4-methylcoumarin-derivatized lysine. This library was used to determine the extended substrate specificities of two trypsin-like enzymes, plasmin and thrombin, which are involved in the blood coagulation pathway. The optimal P4 to P2 substrate specificity for plasmin was P4-Lys/Nle (norleucine)/Val/Ile/Phe, P3-Xaa, and P2-Tyr/Phe/Trp. This cleavage sequence has recently been identified in some of plasmin's physiological substrates. The optimal P4 to P2 extended substrate sequence determined for thrombin was P4-Nle/Leu/Ile/Phe/Val, P3-Xaa, and P2-Pro, a sequence found in many of the physiological substrates of thrombin. Single-substrate kinetic analysis of plasmin and thrombin was used to validate the substrate preferences resulting from the PS-SCL. By three-dimensional structural modeling of the substrates into the active sites of plasmin and thrombin, we identified potential determinants of the defined substrate specificity. This method is amenable to the incorporation of diverse substituents at the P1 position for exploring molecular recognition elements in proteolytic enzymes.
- Published
- 2000
- Full Text
- View/download PDF
35. Chiral N-Acyl-tert-butanesulfinamides: The "Safety-Catch" Principle Applied to Diastereoselective Enolate Alkylations.
- Author
-
Backes BJ, Dragoli DR, and Ellman JA
- Abstract
Diastereoselective enolate alkylation reactions of N-acylsulfinamides and conversion of the alkylation products to a variety of enantiopure products are reported. Several sulfinamides were prepared in solution followed by acylation to provide N-acylsulfinamides. The N-acylsulfinamides were then evaluated in diastereoselective enolate alkylation reactions. Of the sulfinamides evaluated, tert-butanesulfinamide provided the highest diastereoselectivity. To establish the potential utility of sulfinamides as versatile auxiliaries, methods were developed for (1) the racemization-free acylation of tert-butanesulfinamide to prepare optically pure N-acyl-tert-butanesulfinamides, (2) the diastereoselective C-alkylation of N-acyl-tert-butanesulfinamides, (3) the conversion of the N-acyl-tert-butanesulfinamides to the active ester equivalent by N-alkylation and S-oxidation, and (4) the cleavage of the N-alkyl-N-acyl-tert-butanesulfonamides to give chiral alcohol, ester, amide, and carboxylic acid target compounds. These studies provide the groundwork for the development of sulfinamides as dual chiral auxiliaries and linkers for the multistep solid-phase synthesis of enantioenriched compounds.
- Published
- 1999
- Full Text
- View/download PDF
36. Solid support linker strategies.
- Author
-
Backes BJ and Ellman JA
- Subjects
- Indicators and Reagents, Organic Chemistry Phenomena, Photochemistry, Chemistry, Organic
- Abstract
The selection of an appropriate linker is critical to the success of any strategy for the solid-phase synthesis of small molecule libraries. While the primary function of the linker is to covalently attach the initial substrate to the support, innovative strategies have emerged recently in which linkers fulfill important auxiliary roles. These include the cleavage of compounds into solution leaving no trace of the support attachment site, cleavage via cyclization, cleavage by introduction of additional diversity into the structure, and cleavage whereby portions of the compound are sequentially released into solution.
- Published
- 1997
- Full Text
- View/download PDF
37. A reverse-phase HPLC assay for measuring the interaction of polyene macrolide antifungal agents with sterols.
- Author
-
Backes BJ and Rychnovsky SD
- Subjects
- Drug Interactions, Amphotericin B metabolism, Chromatography, Affinity methods, Chromatography, High Pressure Liquid methods, Natamycin metabolism, Sterols metabolism
- Abstract
A quick and simple affinity chromatography method for gauging the interaction of polyene antifungal agents with sterols has been developed. The required affinity columns are prepared from a standard C-18 reverse-phase HPLC column by injecting a measured quantity of sterol under conditions where it is completely retained. After the assay, the sterol is eluted with a less polar solvent and the column reused. By comparing the elution volume of a polyene injected onto the sterol-free column (Ve) with that of the polyene injected onto the sterol-doped column (V), an association constant (Ka) for the polyene-sterol complex was determined. Association constants of different amphotericin B-sterol and pimaricin-sterol complexes were determined and correlated with the polyene's ability to induce membrane permeability and its antifungal properties. This procedure provides a new tool for screening polyene macrolides for antifungal therapy.
- Published
- 1992
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.