1
|
Nascimento M, Moura S, Parra L, Vasconcellos V, Costa G, Leite D, Dias M, Fernandes TVA, Hoelz L, Pimentel L, Bastos M, Boechat N. Ponatinib: A Review of the History of Medicinal Chemistry behind Its Development. Pharmaceuticals (Basel) 2024; 17:1361. [PMID: 39459001 PMCID: PMC11510555 DOI: 10.3390/ph17101361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The primary treatment for chronic myeloid leukemia (CML) involves first- and second-generation tyrosine kinase inhibitors (TKIs), such as imatinib, nilotinib, bosutinib, and dasatinib. However, these medications are ineffective against mutations in the kinase domain of the ABL1 protein, particularly in the protein with the T315I mutation. To address this, ponatinib (PNT), a third-generation inhibitor, was developed. Despite its efficacy in treating the BCR-ABL1T315I mutation, the use of PNT was briefly suspended in 2013 due to serious adverse effects but was subsequently reintroduced to the market. During the drug discovery and development process, it is rare to consolidate all information into a single article, as is the case with ponatinib. This review aims to compile and chronologically organize the research on the discovery of ponatinib using medicinal chemistry tools and computational methods. It includes in silico calculations, such as the octanol/water partition coefficient (cLogP) via SwissAdme, and 2D maps of intermolecular interactions through molecular docking. This approach enhances understanding for both specialists and those interested in medicinal chemistry and pharmacology, while also contextualizing future directions for further optimizations of ponatinib, facilitating the development of new analogs of this crucial inhibitor for the treatment of CML and Philadelphia chromosome-positive acute lymphoblastic leukemia (ALL).
Collapse
Affiliation(s)
- Mayara Nascimento
- Programa de Pós-Graduação em Farmacologia e Química Medicinal do Instituto de Ciências Biomédicas–ICB-UFRJ, Centro de Ciências da Saúde-CCS, Bloco J, Ilha do Fundão, Rio de Janeiro 21941-902, RJ, Brazil; (M.N.); (S.M.)
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| | - Stefany Moura
- Programa de Pós-Graduação em Farmacologia e Química Medicinal do Instituto de Ciências Biomédicas–ICB-UFRJ, Centro de Ciências da Saúde-CCS, Bloco J, Ilha do Fundão, Rio de Janeiro 21941-902, RJ, Brazil; (M.N.); (S.M.)
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| | - Lidia Parra
- Programa de Pós-Graduação Acadêmico em Pesquisa Translacional em Fármacos e Medicamentos–Farmanguinhos, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil;
| | - Valeska Vasconcellos
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
- Programa de Pós-Graduação Acadêmico em Pesquisa Translacional em Fármacos e Medicamentos–Farmanguinhos, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil;
| | - Gabriela Costa
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| | - Debora Leite
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| | - Maria Dias
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| | - Tácio Vinício Amorim Fernandes
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| | - Lucas Hoelz
- Laboratório Computacional de Química Medicinal—LCQM, Instituto Federal do Rio de Janeiro—IFRJ, Campus Pinheiral, Pinheiral 27197-000, RJ, Brazil;
| | - Luiz Pimentel
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| | - Monica Bastos
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| | - Nubia Boechat
- Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos–Fiocruz, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (V.V.); (G.C.); (D.L.); (M.D.); (T.V.A.F.); (L.P.); (M.B.)
| |
Collapse
|
2
|
Xie Q, Liao Q, Wang L, Zhang Y, Chen J, Bai H, Li K, Ai J. The Dominant Mechanism of Cyclophosphamide-Induced Damage to Ovarian Reserve: Premature Activation or Apoptosis of Primordial Follicles? Reprod Sci 2024; 31:30-44. [PMID: 37486531 DOI: 10.1007/s43032-023-01294-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 06/30/2023] [Indexed: 07/25/2023]
Abstract
Cyclophosphamide (CPM), a part of most cancer treatment regimens, has demonstrated high gonadal toxicity in females. Initially, CPM is believed to damage the ovarian reserve by premature activation of primordial follicles, for the fact that facing CPM damage, primordial oocytes show the activation of PTEN/PI3K/AKT pathways, accompanied by accelerated activation of follicle developmental waves. Meanwhile, primordial follicles are dormant and not considered the target of CPM. However, many researchers have found DNA DSBs and apoptosis within primordial oocytes under CPM-induced ovarian damage instead of premature accelerated activation. A stricter surveillance system of DNA damage is also thought to be in primordial oocytes. So far, the apoptotic death mechanism is considered well-proved, but the premature activation theory is controversial and unacceptable. The connection between the upregulation of PTEN/PI3K/AKT pathways and DNA DSBs and apoptosis within primordial oocytes is also unclear. This review aims to highlight the flaw and/or support of the disputed premature activation theory and the apoptosis mechanism to identify the underlying mechanism of CPM's injury on ovarian reserve, which is crucial to facilitate the discovery and development of effective ovarian protectants. Ultimately, this review finds no good evidence for follicle activation and strong consistent evidence for apoptosis.
Collapse
Affiliation(s)
- Qin Xie
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Department of Reproductive Medicine Center, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Road, Xiangcheng District, Xiangyang, 441021, Hubei Province, People's Republic of China
| | - Qiuyue Liao
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lingjuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan Zhang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jing Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hualin Bai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Kezhen Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Jihui Ai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
3
|
Sporbeck K, Haas ML, Pastor-Maldonado CJ, Schüssele DS, Hunter C, Takacs Z, Diogo de Oliveira AL, Franz-Wachtel M, Charsou C, Pfisterer SG, Gubas A, Haller PK, Knorr RL, Kaulich M, Macek B, Eskelinen EL, Simonsen A, Proikas-Cezanne T. The ABL-MYC axis controls WIPI1-enhanced autophagy in lifespan extension. Commun Biol 2023; 6:872. [PMID: 37620393 PMCID: PMC10449903 DOI: 10.1038/s42003-023-05236-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 08/10/2023] [Indexed: 08/26/2023] Open
Abstract
Human WIPI β-propellers function as PI3P effectors in autophagy, with WIPI4 and WIPI3 being able to link autophagy control by AMPK and TORC1 to the formation of autophagosomes. WIPI1, instead, assists WIPI2 in efficiently recruiting the ATG16L1 complex at the nascent autophagosome, which in turn promotes lipidation of LC3/GABARAP and autophagosome maturation. However, the specific role of WIPI1 and its regulation are unknown. Here, we discovered the ABL-ERK-MYC signalling axis controlling WIPI1. As a result of this signalling, MYC binds to the WIPI1 promoter and represses WIPI1 gene expression. When ABL-ERK-MYC signalling is counteracted, increased WIPI1 gene expression enhances the formation of autophagic membranes capable of migrating through tunnelling nanotubes to neighbouring cells with low autophagic activity. ABL-regulated WIPI1 function is relevant to lifespan control, as ABL deficiency in C. elegans increased gene expression of the WIPI1 orthologue ATG-18 and prolonged lifespan in a manner dependent on ATG-18. We propose that WIPI1 acts as an enhancer of autophagy that is physiologically relevant for regulating the level of autophagic activity over the lifespan.
Collapse
Affiliation(s)
- Katharina Sporbeck
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Maximilian L Haas
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Carmen J Pastor-Maldonado
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - David S Schüssele
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Catherine Hunter
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Zsuzsanna Takacs
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- Institute of Molecular Biotechnology, A-1030, Vienna, Austria
| | - Ana L Diogo de Oliveira
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Mirita Franz-Wachtel
- Proteome Center Tübingen, Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Chara Charsou
- Institute of Basic Medical Sciences, University of Oslo, 0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0316, Oslo, Norway
| | - Simon G Pfisterer
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- Department of Anatomy, Faculty of Medicine, University of Helsinki, FI-00290, Helsinki, Finland
| | - Andrea Gubas
- Institute of Biochemistry II, Frankfurt Cancer Institute, Goethe University Medical School, D-60590, Frankfurt, Germany
| | - Patricia K Haller
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Roland L Knorr
- Humboldt University of Berlin, Institute of Biology, D-10115, Berlin, Germany
- Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
- International Research Frontiers Initiative, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, 226-8503, Japan
| | - Manuel Kaulich
- Institute of Biochemistry II, Frankfurt Cancer Institute, Goethe University Medical School, D-60590, Frankfurt, Germany
| | - Boris Macek
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- Proteome Center Tübingen, Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Eeva-Liisa Eskelinen
- Department of Biosciences, University of Helsinki, Fl-00790, Helsinki, Finland
- Institute of Biomedicine, University of Turku, FI-20520, Turku, Finland
| | - Anne Simonsen
- Institute of Basic Medical Sciences, University of Oslo, 0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0316, Oslo, Norway
| | - Tassula Proikas-Cezanne
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany.
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany.
| |
Collapse
|
4
|
Pergu R, Shoba VM, Chaudhary SK, Munkanatta Godage DNP, Deb A, Singha S, Dhawa U, Singh P, Anokhina V, Singh S, Siriwardena SU, Choudhary A. Development and Applications of Chimera Platforms for Tyrosine Phosphorylation. ACS CENTRAL SCIENCE 2023; 9:1558-1566. [PMID: 37637727 PMCID: PMC10450875 DOI: 10.1021/acscentsci.3c00200] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Indexed: 08/29/2023]
Abstract
Chimeric small molecules that induce post-translational modification (PTM) on a target protein by bringing it into proximity to a PTM-inducing enzyme are furnishing novel modalities to perturb protein function. Despite recent advances, such molecules are unavailable for a critical PTM, tyrosine phosphorylation. Furthermore, the contemporary design paradigm of chimeric molecules, formed by joining a noninhibitory binder of the PTM-inducing enzyme with the binder of the target protein, prohibits the recruitment of most PTM-inducing enzymes as their noninhibitory binders are unavailable. Here, we report two platforms to generate phosphorylation-inducing chimeric small molecules (PHICS) for tyrosine phosphorylation. We generate PHICS from both noninhibitory binders (scantily available, platform 1) and kinase inhibitors (abundantly available, platform 2) using cysteine-based group transfer chemistry. PHICS triggered phosphorylation on tyrosine residues in diverse sequence contexts and target proteins (e.g., membrane-associated, cytosolic) and displayed multiple bioactivities, including the initiation of a growth receptor signaling cascade and the death of drug-resistant cancer cells. These studies provide an approach to induce biologically relevant PTM and lay the foundation for pharmacologic PTM editing (i.e., induction or removal) of target proteins using abundantly available inhibitors of PTM-inducing or -erasing enzymes.
Collapse
Affiliation(s)
- Rajaiah Pergu
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Veronika M. Shoba
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Santosh K. Chaudhary
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | | | - Arghya Deb
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Santanu Singha
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Uttam Dhawa
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Prashant Singh
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Viktoriya Anokhina
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Sameek Singh
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Sachini U. Siriwardena
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Amit Choudhary
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Medicine, Harvard Medical School, Boston, Massachusetts 02115, United States
- Divisions
of Renal Medicine and Engineering, Brigham
and Women’s Hospital, Boston, Massachusetts 02115, United States
| |
Collapse
|
5
|
Mazzera L, Abeltino M, Lombardi G, Cantoni AM, Jottini S, Corradi A, Ricca M, Rossetti E, Armando F, Peli A, Ferrari A, Martinelli G, Scupoli MT, Visco C, Bonifacio M, Ripamonti A, Gambacorti-Passerini C, Bonati A, Perris R, Lunghi P. MEK1/2 regulate normal BCR and ABL1 tumor-suppressor functions to dictate ATO response in TKI-resistant Ph+ leukemia. Leukemia 2023; 37:1671-1685. [PMID: 37386079 PMCID: PMC10400427 DOI: 10.1038/s41375-023-01940-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/10/2023] [Accepted: 06/07/2023] [Indexed: 07/01/2023]
Abstract
Resistance to tyrosine kinase inhibitors (TKIs) remains a clinical challenge in Ph-positive variants of chronic myeloid leukemia. We provide mechanistic insights into a previously undisclosed MEK1/2/BCR::ABL1/BCR/ABL1-driven signaling loop that may determine the efficacy of arsenic trioxide (ATO) in TKI-resistant leukemic patients. We find that activated MEK1/2 assemble into a pentameric complex with BCR::ABL1, BCR and ABL1 to induce phosphorylation of BCR and BCR::ABL1 at Tyr360 and Tyr177, and ABL1, at Thr735 and Tyr412 residues thus provoking loss of BCR's tumor-suppression functions, enhanced oncogenic activity of BCR::ABL1, cytoplasmic retention of ABL1 and consequently drug resistance. Coherently, pharmacological blockade of MEK1/2 induces dissociation of the pentameric MEK1/2/BCR::ABL1/BCR/ABL1 complex and causes a concurrent BCRY360/Y177, BCR::ABL1Y360/Y177 and cytoplasmic ABL1Y412/T735 dephosphorylation thereby provoking the rescue of the BCR's anti-oncogenic activities, nuclear accumulation of ABL1 with tumor-suppressive functions and consequently, growth inhibition of the leukemic cells and an ATO sensitization via BCR-MYC and ABL1-p73 signaling axes activation. Additionally, the allosteric activation of nuclear ABL1 was consistently found to enhance the anti-leukemic effects of the MEK1/2 inhibitor Mirdametinib, which when combined with ATO, significantly prolonged the survival of mice bearing BCR::ABL1-T315I-induced leukemia. These findings highlight the therapeutic potential of MEK1/2-inhibitors/ATO combination for the treatment of TKI-resistant leukemia.
Collapse
Affiliation(s)
- Laura Mazzera
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "Bruno Ubertini", Brescia, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Manuela Abeltino
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Guerino Lombardi
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "Bruno Ubertini", Brescia, Italy
| | | | - Stefano Jottini
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Attilio Corradi
- Department of Veterinary Science, University of Parma, Parma, Italy
| | - Micaela Ricca
- Istituto Zooprofilattico Sperimentale della Lombardia e dell'Emilia Romagna "Bruno Ubertini", Brescia, Italy
| | - Elena Rossetti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- National Healthcare Service (SSN-Servizio Sanitario Nazionale) ASL Piacenza, Piacenza, Italy
| | - Federico Armando
- Department of Veterinary Science, University of Parma, Parma, Italy
- University of Veterinary Medicine Hannover, Foundation, Hanover, Germany
| | - Angelo Peli
- Department for Life Quality Studies Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Anna Ferrari
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, FC, Italy
| | - Giovanni Martinelli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, FC, Italy
- Institute of Hematology "L. e A. Seragnoli", Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Maria Teresa Scupoli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Carlo Visco
- Department of Engineering for Innovation Medicine, Section of Hematology-University of Verona, Verona, Italy
| | - Massimiliano Bonifacio
- Department of Engineering for Innovation Medicine, Section of Hematology-University of Verona, Verona, Italy
| | - Alessia Ripamonti
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
- Adult Hematology, IRCCS San Gerardo, Monza, Italy
| | - Carlo Gambacorti-Passerini
- Department of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
- Adult Hematology, IRCCS San Gerardo, Monza, Italy
| | - Antonio Bonati
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Roberto Perris
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Centre for Molecular and Translational Oncology-COMT, University of Parma, Parma, Italy
| | - Paolo Lunghi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy.
- Centre for Molecular and Translational Oncology-COMT, University of Parma, Parma, Italy.
| |
Collapse
|
6
|
Golovine K, Abalakov G, Lian Z, Chatla S, Karami A, Chitrala KN, Madzo J, Nieborowska-Skorska M, Huang J, Skorski T. ABL1 kinase as a tumor suppressor in AML1-ETO and NUP98-PMX1 leukemias. Blood Cancer J 2023; 13:42. [PMID: 36959186 PMCID: PMC10036529 DOI: 10.1038/s41408-023-00810-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/25/2023] Open
Abstract
Deletion of ABL1 was detected in a cohort of hematologic malignancies carrying AML1-ETO and NUP98 fusion proteins. Abl1-/- murine hematopoietic cells transduced with AML1-ETO and NUP98-PMX1 gained proliferation advantage when compared to Abl1 + /+ counterparts. Conversely, overexpression and pharmacological stimulation of ABL1 kinase resulted in reduced proliferation. To pinpoint mechanisms facilitating the transformation of ABL1-deficient cells, Abl1 was knocked down in 32Dcl3-Abl1ko cells by CRISPR/Cas9 followed by the challenge of growth factor withdrawal. 32Dcl3-Abl1ko cells but not 32Dcl3-Abl1wt cells generated growth factor-independent clones. RNA-seq implicated PI3K signaling as one of the dominant mechanisms contributing to growth factor independence in 32Dcl3-Abl1ko cells. PI3K inhibitor buparlisib exerted selective activity against Lin-cKit+ NUP98-PMX1;Abl1-/- cells when compared to the Abl1 + /+ counterparts. Since the role of ABL1 in DNA damage response (DDR) is well established, we also tested the inhibitors of ATM (ATMi), ATR (ATRi) and DNA-PKcs (DNA-PKi). AML1-ETO;Abl1-/- and NUP98-PMX1;Abl1-/- cells were hypersensitive to DNA-PKi and ATRi, respectively, when compared to Abl1 + /+ counterparts. Moreover, ABL1 kinase inhibitor enhanced the sensitivity to PI3K, DNA-PKcs and ATR inhibitors. In conclusion, we showed that ABL1 kinase plays a tumor suppressor role in hematological malignancies induced by AML1-ETO and NUP98-PMX1 and modulates the response to PI3K and/or DDR inhibitors.
Collapse
Affiliation(s)
- Konstantin Golovine
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Gleb Abalakov
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Zhaorui Lian
- Coriell Institute for Medical Research, Camden, NJ, USA
| | - Srinivas Chatla
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Adam Karami
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Kumaraswamy Naidu Chitrala
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Jozef Madzo
- Coriell Institute for Medical Research, Camden, NJ, USA
| | - Margaret Nieborowska-Skorska
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Jian Huang
- Coriell Institute for Medical Research, Camden, NJ, USA.
| | - Tomasz Skorski
- Fels Cancer Institute for Personalized Medicine, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
7
|
Jiang C, He X, Wang Y, Chen CJ, Othman Y, Hao Y, Yuan J, Xie XQ, Feng Z. Molecular Modeling Study of a Receptor-Orthosteric Ligand-Allosteric Modulator Signaling Complex. ACS Chem Neurosci 2023; 14:418-434. [PMID: 36692197 PMCID: PMC10032570 DOI: 10.1021/acschemneuro.2c00554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/09/2023] [Indexed: 01/25/2023] Open
Abstract
Allosteric modulators (AMs) are considered as a perpetual hotspot in research for their higher selectivity and various effects on orthosteric ligands (OL). They are classified in terms of their functionalities as positive, negative, or silent allosteric modulators (PAM, NAM, or SAM, respectively). In the present work, 11 pairs of three-dimensional (3D) structures of receptor-orthosteric ligand and receptor-orthosteric ligand-allosteric modulator complexes have been collected for the studies, including three different systems: GPCR, enzyme, and ion channel. Molecular dynamics (MD) simulations are applied to quantify the dynamic interactions in both the orthosteric and allosteric binding pockets and the structural fluctuation of the involved proteins. Our results showed that MD simulations of moderately large molecules or peptides undergo insignificant changes compared to crystal structure results. Furthermore, we also studied the conformational changes of receptors that bound with PAM and NAM, as well as the different allosteric binding sites in a receptor. There should be no preference for the position of the allosteric binding pocket after comparing the allosteric binding pockets of these three systems. Finally, we aligned four distinct β2 adrenoceptor structures and three N-methyl-d-aspartate receptor (NMDAR) structures to investigate conformational changes. In the β2 adrenoceptor systems, the aligned results revealed that transmembrane (TM) helices 1, 5, and 6 gradually increased outward movement from an enhanced inactive state to an improved active state. TM6 endured the most significant conformational changes (around 11 Å). For NMDAR, the bottom section of NMDAR's ligand-binding domain (LBD) experienced an upward and outward shift during the gradually activating process. In conclusion, our research provides insight into receptor-orthosteric ligand-allosteric modulator studies and the design and development of allosteric modulator drugs using MD simulation.
Collapse
Affiliation(s)
- Chen Jiang
- Department
of Pharmaceutical Sciences and Computational Chemical Genomics Screening
Center, Pharmacometrics & System Pharmacology (PSP) PharmacoAnalytics,
School of Pharmacy; National Center of Excellence for Computational
Drug Abuse Research; Drug Discovery Institute; Departments of Computational
Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania15261, United States
| | - Xibing He
- Department
of Pharmaceutical Sciences and Computational Chemical Genomics Screening
Center, Pharmacometrics & System Pharmacology (PSP) PharmacoAnalytics,
School of Pharmacy; National Center of Excellence for Computational
Drug Abuse Research; Drug Discovery Institute; Departments of Computational
Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania15261, United States
| | - Yuanqiang Wang
- School
of Pharmacy and Bioengineering, Chongqing
University of Technology, Chongqing400054, China
- Chongqing
Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing400054, China
- Chongqing
Key Laboratory of Target Based Drug Screening and Effect Evaluation, Chongqing400054, China
| | - Chih-Jung Chen
- Department
of Pharmaceutical Sciences and Computational Chemical Genomics Screening
Center, Pharmacometrics & System Pharmacology (PSP) PharmacoAnalytics,
School of Pharmacy; National Center of Excellence for Computational
Drug Abuse Research; Drug Discovery Institute; Departments of Computational
Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania15261, United States
| | - Yasmin Othman
- Department
of Pharmaceutical Sciences and Computational Chemical Genomics Screening
Center, Pharmacometrics & System Pharmacology (PSP) PharmacoAnalytics,
School of Pharmacy; National Center of Excellence for Computational
Drug Abuse Research; Drug Discovery Institute; Departments of Computational
Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania15261, United States
| | - Yixuan Hao
- Department
of Pharmaceutical Sciences and Computational Chemical Genomics Screening
Center, Pharmacometrics & System Pharmacology (PSP) PharmacoAnalytics,
School of Pharmacy; National Center of Excellence for Computational
Drug Abuse Research; Drug Discovery Institute; Departments of Computational
Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania15261, United States
| | - Jiayi Yuan
- Department
of Pharmaceutical Sciences and Computational Chemical Genomics Screening
Center, Pharmacometrics & System Pharmacology (PSP) PharmacoAnalytics,
School of Pharmacy; National Center of Excellence for Computational
Drug Abuse Research; Drug Discovery Institute; Departments of Computational
Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania15261, United States
| | - Xiang-Qun Xie
- Department
of Pharmaceutical Sciences and Computational Chemical Genomics Screening
Center, Pharmacometrics & System Pharmacology (PSP) PharmacoAnalytics,
School of Pharmacy; National Center of Excellence for Computational
Drug Abuse Research; Drug Discovery Institute; Departments of Computational
Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania15261, United States
| | - Zhiwei Feng
- Department
of Pharmaceutical Sciences and Computational Chemical Genomics Screening
Center, Pharmacometrics & System Pharmacology (PSP) PharmacoAnalytics,
School of Pharmacy; National Center of Excellence for Computational
Drug Abuse Research; Drug Discovery Institute; Departments of Computational
Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania15261, United States
| |
Collapse
|
8
|
Properties of FDA-approved small molecule protein kinase inhibitors: A 2023 update. Pharmacol Res 2023; 187:106552. [PMID: 36403719 DOI: 10.1016/j.phrs.2022.106552] [Citation(s) in RCA: 146] [Impact Index Per Article: 73.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 11/18/2022]
Abstract
Owing to the dysregulation of protein kinase activity in many diseases including cancer, this enzyme family has become one of the most important drug targets in the 21st century. There are 72 FDA-approved therapeutic agents that target about two dozen different protein kinases and three of these drugs were approved in 2022. Of the approved drugs, twelve target protein-serine/threonine protein kinases, four are directed against dual specificity protein kinases (MEK1/2), sixteen block nonreceptor protein-tyrosine kinases, and 40 target receptor protein-tyrosine kinases. The data indicate that 62 of these drugs are prescribed for the treatment of neoplasms (57 against solid tumors including breast, lung, and colon, ten against nonsolid tumors such as leukemia, and four against both solid and nonsolid tumors: acalabrutinib, ibrutinib, imatinib, and midostaurin). Four drugs (abrocitinib, baricitinib, tofacitinib, upadacitinib) are used for the treatment of inflammatory diseases (atopic dermatitis, psoriatic arthritis, rheumatoid arthritis, Crohn disease, and ulcerative colitis). Of the 72 approved drugs, eighteen are used in the treatment of multiple diseases. The following three drugs received FDA approval in 2022 for the treatment of these specified diseases: abrocitinib (atopic dermatitis), futibatinib (cholangiocarcinomas), pacritinib (myelofibrosis). All of the FDA-approved drugs are orally effective with the exception of netarsudil, temsirolimus, and trilaciclib. This review summarizes the physicochemical properties of all 72 FDA-approved small molecule protein kinase inhibitors including lipophilic efficiency and ligand efficiency.
Collapse
|
9
|
Krishnan K, Tian H, Tao P, Verkhivker GM. Probing conformational landscapes and mechanisms of allosteric communication in the functional states of the ABL kinase domain using multiscale simulations and network-based mutational profiling of allosteric residue potentials. J Chem Phys 2022; 157:245101. [PMID: 36586979 PMCID: PMC11184971 DOI: 10.1063/5.0133826] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
In the current study, multiscale simulation approaches and dynamic network methods are employed to examine the dynamic and energetic details of conformational landscapes and allosteric interactions in the ABL kinase domain that determine the kinase functions. Using a plethora of synergistic computational approaches, we elucidate how conformational transitions between the active and inactive ABL states can employ allosteric regulatory switches to modulate intramolecular communication networks between the ATP site, the substrate binding region, and the allosteric binding pocket. A perturbation-based network approach that implements mutational profiling of allosteric residue propensities and communications in the ABL states is proposed. Consistent with biophysical experiments, the results reveal functionally significant shifts of the allosteric interaction networks in which preferential communication paths between the ATP binding site and substrate regions in the active ABL state become suppressed in the closed inactive ABL form, which in turn features favorable allosteric coupling between the ATP site and the allosteric binding pocket. By integrating the results of atomistic simulations with dimensionality reduction methods and Markov state models, we analyze the mechanistic role of macrostates and characterize kinetic transitions between the ABL conformational states. Using network-based mutational scanning of allosteric residue propensities, this study provides a comprehensive computational analysis of long-range communications in the ABL kinase domain and identifies conserved regulatory hotspots that modulate kinase activity and allosteric crosstalk between the allosteric pocket, ATP binding site, and substrate binding regions.
Collapse
Affiliation(s)
| | - Hao Tian
- Department of Chemistry, Center for Research Computing, Center for Drug Discovery, Design, and Delivery (CD4), Southern Methodist University, Dallas, Texas 75205, USA
| | - Peng Tao
- Department of Chemistry, Center for Research Computing, Center for Drug Discovery, Design, and Delivery (CD4), Southern Methodist University, Dallas, Texas 75205, USA
| | - Gennady M. Verkhivker
- Author to whom correspondence should be addressed: . Telephone: 714-516-4586. Fax: 714-532-6048
| |
Collapse
|
10
|
Asgaonkar K, Tanksali S, Abhang K, Sagar A. Development of optimized pyrimido-thiazole scaffold derivatives as anticancer and multitargeting tyrosine kinase inhibitors using computational studies. J INDIAN CHEM SOC 2022. [DOI: 10.1016/j.jics.2022.100803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
11
|
Teng M, Young DW, Tan Z. The Pursuit of Enzyme Activation: A Snapshot of the Gold Rush. J Med Chem 2022; 65:14289-14304. [PMID: 36265019 DOI: 10.1021/acs.jmedchem.2c01291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A range of enzymes drive human physiology, and their activities are tightly regulated through numerous signaling pathways. Depending on the context, these pathways may activate or inhibit an enzyme as a way to ensure proper execution of cellular functions. From a drug discovery and development perspective, pharmacological inhibition of enzymes has been a focus of interest, as many diseases are associated with the upregulation of enzyme function. On the other hand, however, pharmacological activation of enzymes such as kinases and phosphatases has been of increasing interest. In this review, we discuss seven case studies that highlight pharmacological activation strategy, describe the binding modes and pharmacology of the activators, and comment on how this on-demand activation strategy complements the commonly pursued inhibition strategy, thus jointly enabling bidirectional modulation of specific target of interest. Going forward, we expect activators to play important roles as chemical probes and drug leads.
Collapse
Affiliation(s)
- Mingxing Teng
- Department of Pathology & Immunology, and Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Damian W Young
- Department of Pathology & Immunology, and Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Zhi Tan
- Department of Pathology & Immunology, and Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, Texas 77030, United States
| |
Collapse
|
12
|
Gros K, Matkovič U, Parato G, Miš K, Luin E, Bernareggi A, Sciancalepore M, Marš T, Lorenzon P, Pirkmajer S. Neuronal Agrin Promotes Proliferation of Primary Human Myoblasts in an Age-Dependent Manner. Int J Mol Sci 2022; 23:ijms231911784. [PMID: 36233091 PMCID: PMC9570459 DOI: 10.3390/ijms231911784] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/06/2022] [Accepted: 09/22/2022] [Indexed: 12/02/2022] Open
Abstract
Neuronal agrin, a heparan sulphate proteoglycan secreted by the α-motor neurons, promotes the formation and maintenance of the neuromuscular junction by binding to Lrp4 and activating muscle-specific kinase (MuSK). Neuronal agrin also promotes myogenesis by enhancing differentiation and maturation of myotubes, but its effect on proliferating human myoblasts, which are often considered to be unresponsive to agrin, remains unclear. Using primary human myoblasts, we determined that neuronal agrin induced transient dephosphorylation of ERK1/2, while c-Abl, STAT3, and focal adhesion kinase were unresponsive. Gene silencing of Lrp4 and MuSK markedly reduced the BrdU incorporation, suggesting the functional importance of the Lrp4/MuSK complex for myoblast proliferation. Acute and chronic treatments with neuronal agrin increased the proliferation of human myoblasts in old donors, but they did not affect the proliferation of myoblasts in young donors. The C-terminal fragment of agrin which lacks the Lrp4-binding site and cannot activate MuSK had a similar age-dependent effect, indicating that the age-dependent signalling pathways activated by neuronal agrin involve the Lrp4/MuSK receptor complex as well as an Lrp4/MuSK-independent pathway which remained unknown. Collectively, our results highlight an age-dependent role for neuronal agrin in promoting the proliferation of human myoblasts.
Collapse
Affiliation(s)
- Katarina Gros
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Urška Matkovič
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Giulia Parato
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- The B.R.A.I.N. Centre for Neuroscience, University of Trieste, 34127 Trieste, Italy
| | - Katarina Miš
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Elisa Luin
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- The B.R.A.I.N. Centre for Neuroscience, University of Trieste, 34127 Trieste, Italy
| | - Annalisa Bernareggi
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- The B.R.A.I.N. Centre for Neuroscience, University of Trieste, 34127 Trieste, Italy
| | - Marina Sciancalepore
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- The B.R.A.I.N. Centre for Neuroscience, University of Trieste, 34127 Trieste, Italy
| | - Tomaž Marš
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Paola Lorenzon
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- The B.R.A.I.N. Centre for Neuroscience, University of Trieste, 34127 Trieste, Italy
- Correspondence: (P.L.); (S.P.)
| | - Sergej Pirkmajer
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Correspondence: (P.L.); (S.P.)
| |
Collapse
|
13
|
McKernan CM, Khatri A, Hannigan M, Child J, Chen Q, Mayro B, Snyder D, Nicchitta CV, Pendergast AM. ABL kinases regulate translation in HER2+ cells through Y-box-binding protein 1 to facilitate colonization of the brain. Cell Rep 2022; 40:111268. [PMID: 36044842 PMCID: PMC9472557 DOI: 10.1016/j.celrep.2022.111268] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 06/20/2022] [Accepted: 08/04/2022] [Indexed: 11/27/2022] Open
Abstract
Patients with human epidermal growth factor receptor 2-positive (HER2+/ERBB2) breast cancer often present with brain metastasis. HER2-targeted therapies have not been successful to treat brain metastases in part due to poor blood-brain barrier (BBB) penetrance and emergence of resistance. Here, we report that Abelson (ABL) kinase allosteric inhibitors improve overall survival and impair HER2+ brain metastatic outgrowth in vivo. Mechanistically, ABL kinases phosphorylate the RNA-binding protein Y-box-binding protein 1 (YB-1). ABL kinase inhibition disrupts binding of YB-1 to the ERBB2 mRNA and impairs translation, leading to a profound decrease in HER2 protein levels. ABL-dependent tyrosine phosphorylation of YB-1 promotes HER2 translation. Notably, loss of YB-1 inhibits brain metastatic outgrowth and impairs expression of a subset of ABL-dependent brain metastatic targets. These data support a role for ABL kinases in the translational regulation of brain metastatic targets through YB-1 and offer a therapeutic target for HER2+ brain metastasis patients.
Collapse
Affiliation(s)
- Courtney M McKernan
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Aaditya Khatri
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA; Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
| | - Molly Hannigan
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jessica Child
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Qiang Chen
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Benjamin Mayro
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - David Snyder
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | | | - Ann Marie Pendergast
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
14
|
Teng M, Luskin MR, Cowan-Jacob SW, Ding Q, Fabbro D, Gray NS. The Dawn of Allosteric BCR-ABL1 Drugs: From a Phenotypic Screening Hit to an Approved Drug. J Med Chem 2022; 65:7581-7594. [PMID: 35609336 DOI: 10.1021/acs.jmedchem.2c00373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chronic myeloid leukemia (CML) is driven by the constitutive activity of the BCR-ABL1 fusion oncoprotein. Despite the great success of drugs that target the BCR-ABL1 ATP-binding site in transforming CML into a manageable disease, emerging resistance point mutations impair inhibitor binding, thereby limiting the effectiveness of these drugs. Recently, allosteric inhibitors that interact with the ABL1 myristate-binding site have been shown to awaken an endogenous regulatory mechanism and reset full-length BCR-ABL1 into an inactive assembled state. The discovery and development of these allosteric inhibitors demonstrates an in-depth understanding of the fundamental regulatory mechanisms of kinases. In this review, we illustrate the structural basis of c-ABL1's dynamic regulation of autoinhibition and activation, discuss the discovery of allosteric inhibitors and the characterization of their mechanism of action, present the therapeutic potential of dual binding to delay the development of mutation-driven acquired resistance, and suggest key lessons learned from this program.
Collapse
Affiliation(s)
- Mingxing Teng
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Marlise R Luskin
- Division of Hematologic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, United States
| | - Sandra W Cowan-Jacob
- Novartis Institutes for BioMedical Research, Novartis Campus, Basel CH-4056, Switzerland
| | - Qiang Ding
- Allorion Therapeutics, Guangzhou, Guangdong 511300, China
| | | | - Nathanael S Gray
- Department of Chemical and Systems Biology, ChEM-H, Stanford Cancer Institute, School of Medicine, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
15
|
A new strategy for the rapid identification and validation of direct toxicity targets of psoralen-induced hepatotoxicity. Toxicol Lett 2022; 363:11-26. [PMID: 35597499 DOI: 10.1016/j.toxlet.2022.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 04/14/2022] [Accepted: 05/10/2022] [Indexed: 12/12/2022]
Abstract
The interaction between small-molecule compounds of traditional Chinese medicine and their direct targets is the molecular initiation event, which is the key factor for toxicity efficacy. Psoralen, an active component of Fructus Psoraleae, is toxic to the liver and has various pharmacological properties. Although the mechanism of psoralen-induced hepatotoxicity has been studied, the direct target of psoralen remains unclear. Thus, the aim of this study was to discover direct targets of psoralen. To this end, we initially used proteomics based on drug affinity responsive target stability (DARTS) technology to identify the direct targets of psoralen. Next, we used surface plasmon resonance (SPR) analysis and verified the affinity effect of the 'component-target protein'. This method combines molecular docking technology to explore binding sites between small molecules and proteins. SPR and molecular docking confirmed that psoralen and tyrosine-protein kinase ABL1 could be stably combined. Based on the above experimental results, ABL1 is a potential direct target of psoralen-induced hepatotoxicity. Finally, the targets Nrf2 and mTOR, which are closely related to the hepatotoxicity caused by psoralen, were predicted by integrating proteomics and network pharmacology. The direct target ABL1 is located upstream of Nrf2 and mTOR, Nrf2 can influence the expression of mTOR by affecting the level of reactive oxygen species. Immunofluorescence experiments and western blot results showed that psoralen could affect ROS levels and downstream Nrf2 and mTOR protein changes, whereas the ABL1 inhibitor imatinib and ABL1 agonist DPH could enhance or inhibit this effect. In summary, we speculated that when psoralen causes hepatotoxicity, it acts on the direct target ABL1, resulting in a decrease in Nrf2 expression, an increase in ROS levels and a reduction in mTOR expression, which may cause cell death. We developed a new strategy for predicting and validating the direct targets of psoralen. This strategy identified the toxic target, ABL1, and the potential toxic mechanism of psoralen.
Collapse
|
16
|
Le Vely B, Phan C, Berrebeh N, Thuillet R, Ottaviani M, Chelgham MK, Chaumais MC, Amazit L, Humbert M, Huertas A, Guignabert C, Tu L. Loss of cAbl Tyrosine Kinase in Pulmonary Arterial Hypertension Causes Dysfunction of Vascular Endothelial Cells. Am J Respir Cell Mol Biol 2022; 67:215-226. [PMID: 35550008 DOI: 10.1165/rcmb.2021-0332oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and fatal disease characterized by the dysfunction of pulmonary endothelial cells (ECs) and obstructive vascular remodeling. The non-receptor tyrosine kinase c-Abelson (cAbl) plays central roles in regulating cell-cycle arrest, apoptosis, and senescence after cellular stress. We hypothesized that cAbl is down-activated in experimental and human PAH, thus leading to reduced DNA integrity and angiogenic capacity of pulmonary ECs from PAH patients (PAH-ECs). We found cAbl and phosphorylated cAbl levels to be lower in the endothelium of remodeled pulmonary vessels in the lungs of PAH patients than controls. Similar observations were obtained for the lungs of sugen+hypoxia (SuHx) and monocrotaline (MCT) rats with established pulmonary hypertension. These in situ abnormalities were also replicated in vitro, with cultured PAH-ECs displaying lower cAbl expression and activity and an altered DNA damage response and capacity of tube formation. Downregulation of cAbl by RNA-interference in Control-ECs or its inhibition with dasatinib resulted in genomic instability and the failure to form tubes, whereas upregulation of cAbl with DPH reduced DNA damage and apoptosis in PAH-ECs. Finally, we establish the existence of crosstalk between cAbl and bone morphogenetic protein receptor type II (BMPRII). This work identifies the loss of cAbl signaling as a novel contributor to pulmonary EC dysfunction associated with PAH.
Collapse
Affiliation(s)
- Benjamin Le Vely
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France
| | - Carole Phan
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France
| | - Nihel Berrebeh
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France
| | - Raphaël Thuillet
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France
| | - Mina Ottaviani
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France
| | - Mustapha Kamel Chelgham
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France
| | - Marie-Camille Chaumais
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France.,Université Paris-Saclay Faculté de Pharmacie, 70620, Chatenay-Malabry, France
| | - Larbi Amazit
- Institut Biomédical de Bicêtre, 46657, UMS_44, Villejuif, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France
| | - Marc Humbert
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France.,Assistance Publique - Hopitaux de Paris, 26930, Service de Pneumologie et Soins Intensifs Respiratoires, Le Kremlin-Bicêtre, France
| | - Alice Huertas
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France.,Assistance Publique - Hopitaux de Paris, 26930, Service de Pneumologie et Soins Intensifs Respiratoires, Le Kremlin-Bicêtre, France
| | - Christophe Guignabert
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France
| | - Ly Tu
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France;
| |
Collapse
|
17
|
Qing T, Liu J, Liu F, Mitchell DC, Beresis RT, Gordan JD. Methods to assess small molecule allosteric modulators of the STRAD pseudokinase. Methods Enzymol 2022; 667:427-453. [PMID: 35525550 DOI: 10.1016/bs.mie.2022.03.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
With the increased appreciation of the biological relevance of pseudokinase (PSK) allostery, the broadening of small molecule strategies to target PSK function is of particular importance. We and others have pursued the development of small molecule allosteric modulators of the STRAD pseudokinase by targeting its ATP binding pocket. The purpose of this effort is to modulate the function of the LKB1 tumor suppressor kinase, which exists in a trimer with the STRAD PSK and the adaptor protein MO25. Here we provide detailed guidance regarding the different methods we have used for medium throughput screening to identify STRAD ligands and measure their impact on LKB1 kinase activity. Our experience supports preferential use of direct measurements of LKB1 kinase activity, and demonstrates the limitations of indirect assessment methods in the development trans-acting allosteric modulators.
Collapse
Affiliation(s)
- Tingting Qing
- Chempartner Co, Ltd., Shanghai, China; Chempartner Co, Ltd., South San Francisco, CA, United States
| | - Jin Liu
- Chempartner Co, Ltd., Shanghai, China; Chempartner Co, Ltd., South San Francisco, CA, United States
| | - Fen Liu
- Chempartner Co, Ltd., Shanghai, China; Chempartner Co, Ltd., South San Francisco, CA, United States
| | - Dom C Mitchell
- Division of Hematology Oncology and Quantitative Biosciences Institute, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Richard T Beresis
- Chempartner Co, Ltd., Shanghai, China; Chempartner Co, Ltd., South San Francisco, CA, United States
| | - John D Gordan
- Division of Hematology Oncology and Quantitative Biosciences Institute, University of California, San Francisco (UCSF), San Francisco, CA, United States.
| |
Collapse
|
18
|
Santos C, Pimentel L, Canzian H, Oliveira A, Junior F, Dantas R, Hoelz L, Marinho D, Cunha A, Bastos M, Boechat N. Hybrids of Imatinib with Quinoline: Synthesis, Antimyeloproliferative Activity Evaluation, and Molecular Docking. Pharmaceuticals (Basel) 2022; 15:ph15030309. [PMID: 35337107 PMCID: PMC8950477 DOI: 10.3390/ph15030309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/09/2022] [Accepted: 02/13/2022] [Indexed: 02/01/2023] Open
Abstract
Imatinib (IMT) is the first-in-class BCR-ABL commercial tyrosine kinase inhibitor (TKI). However, the resistance and toxicity associated with the use of IMT highlight the importance of the search for new TKIs. In this context, heterocyclic systems, such as quinoline, which is present as a pharmacophore in the structure of the TKI inhibitor bosutinib (BST), have been widely applied. Thus, this work aimed to obtain new hybrids of imatinib containing quinoline moieties and evaluate them against K562 cells. The compounds were synthesized with a high purity degree. Among the produced molecules, the inhibitor 4-methyl-N3-(4-(pyridin-3-yl)pyrimidin-2-yl)-N1-(quinolin-4-yl)benzene-1,3-diamine (2g) showed a suitable reduction in cell viability, with a CC50 value of 0.9 µM (IMT, CC50 = 0.08 µM). Molecular docking results suggest that the interaction between the most active inhibitor 2g and the BCR-ABL1 enzyme occurs at the bosutinib binding site through a competitive inhibition mechanism. Despite being less potent and selective than IMT, 2g is a suitable prototype for use in the search for new drugs against chronic myeloid leukemia (CML), especially in patients with acquired resistance to IMT.
Collapse
Affiliation(s)
- Carine Santos
- Laboratório de Sintese de Farmacos-LASFAR, Instituto de Tecnologia em Farmacos-Farmanguinhos, FIOCRUZ, Rua Sizenando Nabuco 100, Manguinhos, Rio de Janeiro 21041-250, Brazil; (C.S.); (L.P.); (H.C.); (A.O.); (L.H.); (D.M.); (M.B.)
- Programa de Pós-graduação em Farmacologia e Química Medicinal do Instituto de Ciências Biomédicas–ICB-UFRJ, Centro de Ciências da Saúde-CCS, Bloco J, Ilha do Fundão, Rio de Janeiro 21941-902, Brazil
| | - Luiz Pimentel
- Laboratório de Sintese de Farmacos-LASFAR, Instituto de Tecnologia em Farmacos-Farmanguinhos, FIOCRUZ, Rua Sizenando Nabuco 100, Manguinhos, Rio de Janeiro 21041-250, Brazil; (C.S.); (L.P.); (H.C.); (A.O.); (L.H.); (D.M.); (M.B.)
| | - Henayle Canzian
- Laboratório de Sintese de Farmacos-LASFAR, Instituto de Tecnologia em Farmacos-Farmanguinhos, FIOCRUZ, Rua Sizenando Nabuco 100, Manguinhos, Rio de Janeiro 21041-250, Brazil; (C.S.); (L.P.); (H.C.); (A.O.); (L.H.); (D.M.); (M.B.)
| | - Andressa Oliveira
- Laboratório de Sintese de Farmacos-LASFAR, Instituto de Tecnologia em Farmacos-Farmanguinhos, FIOCRUZ, Rua Sizenando Nabuco 100, Manguinhos, Rio de Janeiro 21041-250, Brazil; (C.S.); (L.P.); (H.C.); (A.O.); (L.H.); (D.M.); (M.B.)
- Programa de Pós-graduação em Farmacologia e Química Medicinal do Instituto de Ciências Biomédicas–ICB-UFRJ, Centro de Ciências da Saúde-CCS, Bloco J, Ilha do Fundão, Rio de Janeiro 21941-902, Brazil
| | - Floriano Junior
- Laboratório de Bioquímica Experimental e Computacional de Fármacos, Instituto Oswaldo Cruz FIOCRUZ, Av. Brasil 4365, Manguinhos, Rio de Janeiro 21040-360, Brazil; (F.J.); (R.D.)
| | - Rafael Dantas
- Laboratório de Bioquímica Experimental e Computacional de Fármacos, Instituto Oswaldo Cruz FIOCRUZ, Av. Brasil 4365, Manguinhos, Rio de Janeiro 21040-360, Brazil; (F.J.); (R.D.)
| | - Lucas Hoelz
- Laboratório de Sintese de Farmacos-LASFAR, Instituto de Tecnologia em Farmacos-Farmanguinhos, FIOCRUZ, Rua Sizenando Nabuco 100, Manguinhos, Rio de Janeiro 21041-250, Brazil; (C.S.); (L.P.); (H.C.); (A.O.); (L.H.); (D.M.); (M.B.)
| | - Debora Marinho
- Laboratório de Sintese de Farmacos-LASFAR, Instituto de Tecnologia em Farmacos-Farmanguinhos, FIOCRUZ, Rua Sizenando Nabuco 100, Manguinhos, Rio de Janeiro 21041-250, Brazil; (C.S.); (L.P.); (H.C.); (A.O.); (L.H.); (D.M.); (M.B.)
| | - Anna Cunha
- Departamento de Química Orgânica, Campus do Valonguinho, Universidade Federal Fluminense–UFF, Niterói 24020-150, Brazil;
| | - Monica Bastos
- Laboratório de Sintese de Farmacos-LASFAR, Instituto de Tecnologia em Farmacos-Farmanguinhos, FIOCRUZ, Rua Sizenando Nabuco 100, Manguinhos, Rio de Janeiro 21041-250, Brazil; (C.S.); (L.P.); (H.C.); (A.O.); (L.H.); (D.M.); (M.B.)
- Programa de Pós-graduação em Farmacologia e Química Medicinal do Instituto de Ciências Biomédicas–ICB-UFRJ, Centro de Ciências da Saúde-CCS, Bloco J, Ilha do Fundão, Rio de Janeiro 21941-902, Brazil
| | - Nubia Boechat
- Laboratório de Sintese de Farmacos-LASFAR, Instituto de Tecnologia em Farmacos-Farmanguinhos, FIOCRUZ, Rua Sizenando Nabuco 100, Manguinhos, Rio de Janeiro 21041-250, Brazil; (C.S.); (L.P.); (H.C.); (A.O.); (L.H.); (D.M.); (M.B.)
- Programa de Pós-graduação em Farmacologia e Química Medicinal do Instituto de Ciências Biomédicas–ICB-UFRJ, Centro de Ciências da Saúde-CCS, Bloco J, Ilha do Fundão, Rio de Janeiro 21941-902, Brazil
- Correspondence: ; Tel.: +55-(21)-3977-2465
| |
Collapse
|
19
|
Targeting BCR-Abl in the treatment of Philadelphia-chromosome positive chronic myelogenous leukemia. Pharmacol Res 2022; 178:106156. [DOI: 10.1016/j.phrs.2022.106156] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/02/2022] [Indexed: 02/07/2023]
|
20
|
Xie T, Saleh T, Rossi P, Miller D, Kalodimos CG. Imatinib can act as an Allosteric Activator of Abl Kinase. J Mol Biol 2022; 434:167349. [PMID: 34774565 PMCID: PMC8752476 DOI: 10.1016/j.jmb.2021.167349] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/15/2021] [Accepted: 11/05/2021] [Indexed: 02/01/2023]
Abstract
Imatinib is an ATP-competitive inhibitor of Bcr-Abl kinase and the first drug approved for chronic myelogenous leukemia (CML) treatment. Here we show that imatinib binds to a secondary, allosteric site located in the myristoyl pocket of Abl to function as an activator of the kinase activity. Abl transitions between an assembled, inhibited state and an extended, activated state. The equilibrium is regulated by the conformation of the αΙ helix, which is located nearby the allosteric pocket. Imatinib binding to the allosteric pocket elicits an αΙ helix conformation that is not compatible with the assembled state, thereby promoting the extended state and stimulating the kinase activity. Although in wild-type Abl the catalytic pocket has a much higher affinity for imatinib than the allosteric pocket does, the two binding affinities are comparable in Abl variants carrying imatinib-resistant mutations in the catalytic site. A previously isolated imatinib-resistant mutation in patients appears to be mediating its function by increasing the affinity of imatinib for the allosteric pocket, providing a hitherto unknown mechanism of drug resistance. Our results highlight the benefit of combining imatinib with allosteric inhibitors to maximize their inhibitory effect on Bcr-Abl.
Collapse
Affiliation(s)
- Tao Xie
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, United States
| | - Tamjeed Saleh
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, United States
| | - Paolo Rossi
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, United States
| | - Darcie Miller
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, United States
| | - Charalampos G Kalodimos
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, United States.
| |
Collapse
|
21
|
Axtman AD. Characterizing the role of the dark kinome in neurodegenerative disease - A mini review. Biochim Biophys Acta Gen Subj 2021; 1865:130014. [PMID: 34547390 DOI: 10.1016/j.bbagen.2021.130014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/26/2021] [Accepted: 09/14/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Drugs that modulate previously unexplored targets could potentially slow or halt the progression of neurodegenerative diseases. Several candidate proteins lie within the dark kinome, those human kinases that have not been well characterized. Much of the kinome (~80%) remains poorly studied, and these targets likely harbor untapped biological potential. SCOPE OF REVIEW This review highlights the significance of kinases as mediators of aberrant pathways in neurodegeneration and provides examples of published high-quality small molecules that modulate some of these kinases. MAJOR CONCLUSIONS There is a need for continued efforts to develop high-quality chemical tools to illuminate the function of understudied kinases in the brain. Potent and selective small molecules enable accurate pairing of an observed phenotype with a protein target. GENERAL SIGNIFICANCE The examples discussed herein support the premise that validation of therapeutic hypotheses surrounding kinase targets can be accomplished via small molecules and they can serve as the basis for disease-focused drug development campaigns.
Collapse
Affiliation(s)
- Alison D Axtman
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Structural Genomics Consortium, Chapel Hill, NC, USA.
| |
Collapse
|
22
|
Pimentel LCF, Hoelz LVB, Canzian HF, Branco FSC, de Oliveira AP, Campos VR, Júnior FPS, Dantas RF, Resende JALC, Cunha AC, Boechat N, Bastos MM. (Phenylamino)pyrimidine-1,2,3-triazole derivatives as analogs of imatinib: searching for novel compounds against chronic myeloid leukemia. Beilstein J Org Chem 2021; 17:2260-2269. [PMID: 34621389 PMCID: PMC8450943 DOI: 10.3762/bjoc.17.144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/19/2021] [Indexed: 11/23/2022] Open
Abstract
The enzyme tyrosine kinase BCR-Abl-1 is the main molecular target in the treatment of chronic myeloid leukemia and can be competitively inhibited by tyrosine kinase inhibitors such as imatinib. New potential competitive inhibitors were synthesized using the (phenylamino)pyrimidine-pyridine (PAPP) group as a pharmacophoric fragment, and these compounds were biologically evaluated. The synthesis of twelve new compounds was performed in three steps and assisted by microwave irradiation in a 1,3-dipolar cycloaddition to obtain 1,2,3-triazole derivatives substituted on carbon C-4 of the triazole nucleus. All compounds were evaluated for their inhibitory activities against a chronic myeloid leukemia cell line (K562) that expresses the enzyme tyrosine kinase BCR-Abl-1 and against healthy cells (WSS-1) to observe their selectivity. Three compounds showed promising results, with IC50 values between 1.0 and 7.3 μM, and were subjected to molecular docking studies. The results suggest that such compounds can interact at the same binding site as imatinib, probably sharing a competitive inhibition mechanism. One compound showed the greatest interaction affinity for BCR-Abl-1 in the docking studies.
Collapse
Affiliation(s)
- Luiz Claudio Ferreira Pimentel
- Laboratorio de Sintese de Farmacos – LASFAR, Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos –Manguinhos, CEP 21041-250, Rio de Janeiro, Brazil
| | - Lucas Villas Boas Hoelz
- Laboratorio de Sintese de Farmacos – LASFAR, Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos –Manguinhos, CEP 21041-250, Rio de Janeiro, Brazil
| | - Henayle Fernandes Canzian
- Laboratorio de Sintese de Farmacos – LASFAR, Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos –Manguinhos, CEP 21041-250, Rio de Janeiro, Brazil
| | - Frederico Silva Castelo Branco
- Laboratorio de Sintese de Farmacos – LASFAR, Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos –Manguinhos, CEP 21041-250, Rio de Janeiro, Brazil
| | - Andressa Paula de Oliveira
- Laboratorio de Sintese de Farmacos – LASFAR, Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos –Manguinhos, CEP 21041-250, Rio de Janeiro, Brazil
| | - Vinicius Rangel Campos
- Departamento de Química Orgânica, Universidade Federal Fluminense, Campus do Valonguinho, CEP 24020-150,Niterói, Brazil
| | - Floriano Paes Silva Júnior
- Laboratório de Bioquímica Experimental e Computacional de Farmacos, Fundaçao Oswaldo Cruz, Instituto Oswaldo Cruz, CEP 21040-900, Rio de Janeiro, Brazil
| | - Rafael Ferreira Dantas
- Laboratório de Bioquímica Experimental e Computacional de Farmacos, Fundaçao Oswaldo Cruz, Instituto Oswaldo Cruz, CEP 21040-900, Rio de Janeiro, Brazil
| | | | - Anna Claudia Cunha
- Departamento de Química Orgânica, Universidade Federal Fluminense, Campus do Valonguinho, CEP 24020-150,Niterói, Brazil
| | - Nubia Boechat
- Laboratorio de Sintese de Farmacos – LASFAR, Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos –Manguinhos, CEP 21041-250, Rio de Janeiro, Brazil
| | - Mônica Macedo Bastos
- Laboratorio de Sintese de Farmacos – LASFAR, Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos –Manguinhos, CEP 21041-250, Rio de Janeiro, Brazil
| |
Collapse
|
23
|
Fernandes PO, Martins DM, de Souza Bozzi A, Martins JPA, de Moraes AH, Maltarollo VG. Molecular insights on ABL kinase activation using tree-based machine learning models and molecular docking. Mol Divers 2021; 25:1301-1314. [PMID: 34191245 PMCID: PMC8241884 DOI: 10.1007/s11030-021-10261-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/18/2021] [Indexed: 12/14/2022]
Abstract
Abelson kinase (c-Abl) is a non-receptor tyrosine kinase involved in several biological processes essential for cell differentiation, migration, proliferation, and survival. This enzyme's activation might be an alternative strategy for treating diseases such as neutropenia induced by chemotherapy, prostate, and breast cancer. Recently, a series of compounds that promote the activation of c-Abl has been identified, opening a promising ground for c-Abl drug development. Structure-based drug design (SBDD) and ligand-based drug design (LBDD) methodologies have significantly impacted recent drug development initiatives. Here, we combined SBDD and LBDD approaches to characterize critical chemical properties and interactions of identified c-Abl's activators. We used molecular docking simulations combined with tree-based machine learning models-decision tree, AdaBoost, and random forest to understand the c-Abl activators' structural features required for binding to myristoyl pocket, and consequently, to promote enzyme and cellular activation. We obtained predictive and robust models with Matthews correlation coefficient values higher than 0.4 for all endpoints and identified characteristics that led to constructing a structure-activity relationship model (SAR).
Collapse
Affiliation(s)
- Philipe Oliveira Fernandes
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Diego Magno Martins
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Aline de Souza Bozzi
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - João Paulo A Martins
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Adolfo Henrique de Moraes
- Departamento de Química, Instituto de Ciências Exatas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vinícius Gonçalves Maltarollo
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil.
| |
Collapse
|
24
|
Duan JL, He HQ, Yu Y, Liu T, Ma SJ, Li F, Jiang YS, Lin X, Li DD, Lv QZ, Ma HH, Jia XM. E3 ligase c-Cbl regulates intestinal inflammation through suppressing fungi-induced noncanonical NF-κB activation. SCIENCE ADVANCES 2021; 7:7/19/eabe5171. [PMID: 33962939 PMCID: PMC8104877 DOI: 10.1126/sciadv.abe5171] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 03/19/2021] [Indexed: 05/08/2023]
Abstract
Intestinal fungi are critical for modulating host immune homeostasis and underlying mechanisms remain unclear. We show that dendritic cell (DC)-specific deficiency of casitas B-lineage lymphoma (c-Cbl) renders mice susceptible to dextran sodium sulfate (DSS)-induced colitis. Mechanistically, we identify that c-Cbl functions downstream of Dectin-2 and Dectin-3 to mediate the ubiquitination and degradation of noncanonical nuclear factor κB subunit RelB. Thus, c-Cbl deficiency in DCs promotes α-mannan-induced activation of RelB, which suppresses p65-mediated transcription of an anti-inflammatory cytokine gene, il10, thereby aggravating DSS-induced colitis. Moreover, suppressing fungal growth with fluconazole or inhibition of RelB activation in vivo attenuates colitis in mice with DC-specific deletion of c-Cbl. We also demonstrate an interaction between c-Cbl and c-Abl tyrosine kinase and find that treatment with DPH, a c-Abl agonist, synergistically increases fungi-induced c-Cbl activation to restrict colitis. Together, these findings unravel a previously unidentified fungi-induced c-Cbl/RelB axis that sustains intestinal homeostasis and protects against intestinal inflammation.
Collapse
Affiliation(s)
- Jie-Lin Duan
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Hui-Qian He
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Yao Yu
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Tao Liu
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Shu-Jun Ma
- Department of Dermatology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Fan Li
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Yan-Shan Jiang
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Xin Lin
- Institute for Immunology, Tsinghua University School of Medicine, Tsinghua University-Peking University Jointed Center for Life Sciences, Beijing 100084, China
| | - De-Dong Li
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Quan-Zhen Lv
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Hui-Hui Ma
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200092, China
| | - Xin-Ming Jia
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200092, China
| |
Collapse
|
25
|
Reichenstein M, Borovok N, Sheinin A, Brider T, Michaelevski I. Abelson Kinases Mediate the Depression of Spontaneous Synaptic Activity Induced by Amyloid Beta 1-42 Peptides. Cell Mol Neurobiol 2021; 41:431-448. [PMID: 32399753 DOI: 10.1007/s10571-020-00858-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 04/27/2020] [Indexed: 10/24/2022]
Abstract
Amyloid beta (Aβ) peptides represent one of the most studied etiological factors of Alzheimer's disease. Nevertheless, the effects elicited by different molecular forms of amyloid beta peptides widely vary between the studies, mostly depending on experimental conditions. Despite the enormous amount of accumulated evidences concerning the pathological effects of amyloid beta peptides, the exact identity of the amyloid beta species is still controversial, and even less is clear as regards to the downstream effectors that mediate the devastating impact of these peptides on synapses in the central nervous system. Recent publications indicate that some of the neurotoxic effects of amyloid beta peptides may be mediated via the activation of proteins belonging to the Abelson non-receptor tyrosine kinase (Abl) family, that are known to regulate actin cytoskeleton structure as well as phosphorylate microtubule-associated tau protein, a hallmark of Alzheimer's disease. By performing series of miniature excitatory postsynaptic currents (mEPSC) recordings in cultured hippocampal cells, we demonstrate that activation of Abl kinases by acute application of 42 amino acid-length monomeric amyloid beta (Aβ1-42) peptides reduces spontaneous synaptic release, while this effect can be rescued by pharmacologic inhibition of Abl kinase activity, or by reduction of Abl expression with small interfering RNAs. Our electrophysiological data are further reinforced by a subsequent biochemical analysis, showing enhanced phosphorylation of Abl kinase substrate CT10 Regulator of Kinase-homolog-Like (Crkl) upon treatment of hippocampal neurons with Aβ peptides. Thus, we conclude that Abl kinase activation may be involved in Aβ-induced weakening of synaptic transmission.
Collapse
Affiliation(s)
- M Reichenstein
- Dept. of Biochemistry and Molecular Biology, Tel Aviv University, 69978, Tel Aviv, Israel
| | - N Borovok
- Dept. of Biochemistry and Molecular Biology, Tel Aviv University, 69978, Tel Aviv, Israel
| | - A Sheinin
- Dept. of Biochemistry and Molecular Biology, Tel Aviv University, 69978, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, 69978, Tel Aviv, Israel
| | - T Brider
- Department of Molecular Biology, Ariel University, 40700, Ariel, Israel
| | - I Michaelevski
- Department of Molecular Biology, Ariel University, 40700, Ariel, Israel.
- Integrative Brain Science Center Ariel, IBSCA, Ariel University, 40700, Ariel, Israel.
- The Adelson Medical School, Ariel University, 40700, Ariel, Israel.
| |
Collapse
|
26
|
Montecino F, González N, Blanco N, Ramírez MJ, González-Martín A, Alvarez AR, Olguín H. c-Abl Kinase Is Required for Satellite Cell Function Through Pax7 Regulation. Front Cell Dev Biol 2021; 9:606403. [PMID: 33777928 PMCID: PMC7990767 DOI: 10.3389/fcell.2021.606403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 02/08/2021] [Indexed: 01/06/2023] Open
Abstract
Satellite cells (SCs) are tissue-specific stem cells responsible for adult skeletal muscle regeneration and maintenance. SCs function is critically dependent on two families of transcription factors: the paired box (Pax) involved in specification and maintenance and the Muscle Regulatory Factors (MRFs), which orchestrate myogenic commitment and differentiation. In turn, signaling events triggered by extrinsic and intrinsic stimuli control their function via post-translational modifications, including ubiquitination and phosphorylation. In this context, the Abelson non-receptor tyrosine kinase (c-Abl) mediates the activation of the p38 α/β MAPK pathway, promoting myogenesis. c-Abl also regulates the activity of the transcription factor MyoD during DNA-damage stress response, pausing differentiation. However, it is not clear if c-Abl modulates other key transcription factors controlling SC function. This work aims to determine the role of c-Abl in SCs myogenic capacity via loss of function approaches in vitro and in vivo. Here we show that c-Abl inhibition or deletion results in a down-regulation of Pax7 mRNA and protein levels, accompanied by decreased Pax7 transcriptional activity, without a significant effect on MRF expression. Additionally, we provide data indicating that Pax7 is directly phosphorylated by c-Abl. Finally, SC-specific c-Abl ablation impairs muscle regeneration upon acute injury. Our results indicate that c-Abl regulates myogenic progression in activated SCs by controlling Pax7 function and expression.
Collapse
Affiliation(s)
- Fabián Montecino
- Laboratory of Tissue Repair and Adult Stem Cells, Department of Molecular and Cell Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Natalia González
- Laboratory of Tissue Repair and Adult Stem Cells, Department of Molecular and Cell Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Natasha Blanco
- Laboratory of Tissue Repair and Adult Stem Cells, Department of Molecular and Cell Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Manuel J Ramírez
- Laboratory of Tissue Repair and Adult Stem Cells, Department of Molecular and Cell Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Adrián González-Martín
- CARE-UC, Department of Molecular and Cell Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandra R Alvarez
- CARE-UC, Department of Molecular and Cell Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Hugo Olguín
- Laboratory of Tissue Repair and Adult Stem Cells, Department of Molecular and Cell Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
27
|
Laufkötter O, Hu H, Miljković F, Bajorath J. Structure- and Similarity-Based Survey of Allosteric Kinase Inhibitors, Activators, and Closely Related Compounds. J Med Chem 2021; 65:922-934. [PMID: 33476146 DOI: 10.1021/acs.jmedchem.0c02076] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Allosteric kinase inhibitors are thought to have high selectivity and are prime candidates for kinase drug discovery. In addition, the exploration of allosteric mechanisms represents an attractive topic for basic research and drug design. Although the identification and characterization of allosteric kinase inhibitors is still far from being routine, X-ray structures of kinase complexes have been determined for a significant number of such inhibitors. On the basis of structural data, allosteric inhibitors can be confirmed. We report a comprehensive survey of allosteric kinase inhibitors and activators from publicly available X-ray structures, map their binding sites, and determine their distribution over binding pockets in kinases. In addition, we discuss structural features of these compounds and identify active structural analogues and high-confidence target annotations, indicating additional activities for a subset of allosteric inhibitors. This contribution aims to provide a detailed structure-based view of allosteric kinase inhibition.
Collapse
Affiliation(s)
- Oliver Laufkötter
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Friedrich-Hirzebruch-Allee 6, D-53115 Bonn, Germany
| | - Huabin Hu
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Friedrich-Hirzebruch-Allee 6, D-53115 Bonn, Germany
| | - Filip Miljković
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Friedrich-Hirzebruch-Allee 6, D-53115 Bonn, Germany
| | - Jürgen Bajorath
- Department of Life Science Informatics, B-IT, LIMES Program Unit Chemical Biology and Medicinal Chemistry, Rheinische Friedrich-Wilhelms-Universität, Friedrich-Hirzebruch-Allee 6, D-53115 Bonn, Germany
| |
Collapse
|
28
|
Intermolecular Interactions in Crystal Structures of Imatinib-Containing Compounds. Int J Mol Sci 2020; 21:ijms21238970. [PMID: 33255944 PMCID: PMC7731260 DOI: 10.3390/ijms21238970] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
Imatinib, one of the most used therapeutic agents to treat leukemia, is an inhibitor that specifically blocks the activity of tyrosine kinases. The molecule of imatinib is flexible and contains several functional groups able to take part in H-bonding and hydrophobic interactions. Analysis of molecular conformations for this drug was carried out using density functional theory calculations of rotation potentials along single bonds and by analyzing crystal structures of imatinib-containing compounds taken from the Cambridge Structural Database and the Protein Data Bank. Rotation along the N-C bond in the region of the amide group was found to be the reason for two relatively stable molecular conformations, an extended and a folded one. The role of various types of intermolecular interactions in stabilization of the particular molecular conformation was studied in terms of (i) the likelihood of H-bond formation, and (ii) their contribution to the Voronoi molecular surface. It is shown that experimentally observed hydrogen bonds are in accord with the likelihood of their formation. The number of H-bonds in ligand-receptor complexes surpasses that in imatinib salts due to the large number of donors and acceptors of H-bonding within the binding pocket of tyrosine kinases. Contribution of hydrophilic intermolecular interactions to the Voronoi molecular surface is similar for both conformations, while π...π stacking is more typical for the folded conformation of imatinib.
Collapse
|
29
|
Sun S, Li L, Wu X, Tang R, Lei C, Wang HH, Huang Y, Nie Z, Yao S. Dual-Product Synergistically Enhanced Colorimetric Assay for Sensitive Detection of Lipid Transferase Activity. Anal Chem 2020; 92:15236-15243. [PMID: 33140958 DOI: 10.1021/acs.analchem.0c03973] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Lipid transferase-catalyzed protein lipidation plays critical roles in many physiological processes and it has been an increasingly attractive therapeutic target from cancer to neurodegeneration, while sensitive detection of lipid transferase activity in biological samples remains challenging. Here, we presented an AuNP-based colorimetric method with dual-product synergistically enhanced sensitivity for convenient detection of lipid transferase activity. Homo sapiens N-myristoyltransferase 1 (HsNMT1), a key lipid transferase, was selected as the model. Accordingly, positively charged substrate peptides (Pep) of HsNMT1 can induce the aggregation of AuNPs through disrupting their electrostatic repulsion, while the HsNMT1-catalyzed lipid modification generates aggregated lipidated peptides (C14-Pep) and negatively charged HS-CoA, which will eliminate the disruption and stabilize the AuNPs by the formation of Au-S bonds, respectively. Consequently, charge reversal of the biomolecules and the formation of Au-S bonds synergistically contribute to the stability of AuNPs in the presence of HsNMT1. Therefore, the HsNMT1 activity can be visually detected by the naked eye through the color change of the AuNPs originated from the change in their distance-dependent surface plasmon resonance absorptions. Here, the A520/A610 ratio can sensitively reflect the activity of HsNMT1 in the linear range of 2-75 nM with a low detection limit of 0.56 nM. Moreover, the method was successfully applied for probing the HsNMT1 activities in different cell lysates and inhibitor screening. Furthermore, given the replaceability of the substrate peptide, the proposed assay is promising for universal application to other lipid transferases and exhibits great potential in lipid transferase-targeted drug development.
Collapse
Affiliation(s)
- Sujuan Sun
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| | - Liangwen Li
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| | - Xianhua Wu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| | - Rui Tang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| | - Chunyang Lei
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| | - Hong-Hui Wang
- College of Biology, Hunan University, Changsha 410082, P. R. China
| | - Yan Huang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| | - Zhou Nie
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| | - Shouzhuo Yao
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan Provincial Key Laboratory of Biomacromolecular Chemical Biology, Hunan University, Changsha 410082, P. R. China
| |
Collapse
|
30
|
Manley PW, Barys L, Cowan-Jacob SW. The specificity of asciminib, a potential treatment for chronic myeloid leukemia, as a myristate-pocket binding ABL inhibitor and analysis of its interactions with mutant forms of BCR-ABL1 kinase. Leuk Res 2020; 98:106458. [DOI: 10.1016/j.leukres.2020.106458] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/22/2020] [Accepted: 09/24/2020] [Indexed: 12/26/2022]
|
31
|
Contreras PS, Tapia PJ, González-Hódar L, Peluso I, Soldati C, Napolitano G, Matarese M, Heras ML, Valls C, Martinez A, Balboa E, Castro J, Leal N, Platt FM, Sobota A, Winter D, Klein AD, Medina DL, Ballabio A, Alvarez AR, Zanlungo S. c-Abl Inhibition Activates TFEB and Promotes Cellular Clearance in a Lysosomal Disorder. iScience 2020; 23:101691. [PMID: 33163944 PMCID: PMC7607485 DOI: 10.1016/j.isci.2020.101691] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/11/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
The transcription factor EB (TFEB) has emerged as a master regulator of lysosomal biogenesis, exocytosis, and autophagy, promoting the clearance of substrates stored in cells. c-Abl is a tyrosine kinase that participates in cellular signaling in physiological and pathophysiological conditions. In this study, we explored the connection between c-Abl and TFEB. Here, we show that under pharmacological and genetic c-Abl inhibition, TFEB translocates into the nucleus promoting the expression of its target genes independently of its well-known regulator, mammalian target of rapamycin complex 1. Active c-Abl induces TFEB phosphorylation on tyrosine and the inhibition of this kinase promotes lysosomal biogenesis, autophagy, and exocytosis. c-Abl inhibition in Niemann-Pick type C (NPC) models, a neurodegenerative disease characterized by cholesterol accumulation in lysosomes, promotes a cholesterol-lowering effect in a TFEB-dependent manner. Thus, c-Abl is a TFEB regulator that mediates its tyrosine phosphorylation, and the inhibition of c-Abl activates TFEB promoting cholesterol clearance in NPC models. c-Abl is a TFEB regulator that mediates its tyr phosphorylation c-Abl inhibition promotes TFEB activity independently of mTORC1 c-Abl inhibition reduces cholesterol accumulation in NPC1 models
Collapse
Affiliation(s)
- Pablo S Contreras
- Department of Cell & Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Alameda 340, Santiago 8331010, Chile.,CARE UC Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Alameda 340, Santiago 8331010, Chile
| | - Pablo J Tapia
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Alameda 340, Santiago 8331010, Chile
| | - Lila González-Hódar
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Alameda 340, Santiago 8331010, Chile
| | - Ivana Peluso
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy
| | - Chiara Soldati
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy
| | - Gennaro Napolitano
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy
| | - Maria Matarese
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy
| | - Macarena Las Heras
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Alameda 340, Santiago 8331010, Chile
| | - Cristian Valls
- Department of Cell & Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Alameda 340, Santiago 8331010, Chile.,CARE UC Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis Martinez
- Department of Cell & Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Alameda 340, Santiago 8331010, Chile.,CARE UC Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Elisa Balboa
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Alameda 340, Santiago 8331010, Chile
| | - Juan Castro
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Alameda 340, Santiago 8331010, Chile
| | - Nancy Leal
- Department of Cell & Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Alameda 340, Santiago 8331010, Chile.,CARE UC Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, Oxford, UK
| | - Andrzej Sobota
- Department of Cell Biology, Nencki Institute of Experimental Biology, 3 Pasteur St., 02-093 Warsaw, Poland
| | - Dominic Winter
- Institute for Biochemistry and Molecular Biology, Rheinische-Friedrich-Wilhelms-University, Bonn, Germany
| | - Andrés D Klein
- Centro de Genética y Genómica, Universidad Del Desarrollo Clínica Alemana de Santiago, Chile
| | - Diego L Medina
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy.,Medical Genetics, Department of Pediatrics, Federico II University, Via Pansini 5, 80131 Naples, Italy.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Alejandra R Alvarez
- Department of Cell & Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Alameda 340, Santiago 8331010, Chile.,CARE UC Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Silvana Zanlungo
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Alameda 340, Santiago 8331010, Chile
| |
Collapse
|
32
|
Signalling input from divergent pathways subverts B cell transformation. Nature 2020; 583:845-851. [PMID: 32699415 PMCID: PMC7394729 DOI: 10.1038/s41586-020-2513-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 04/28/2020] [Indexed: 01/29/2023]
Abstract
Malignant transformation typically involves multiple genetic lesions whose combined activity gives rise to cancer1. Our analysis of 1,148 patient-derived B-cell leukemia (B-ALL) samples revealed that individual mutations did not promote leukemogenesis unless they converged on one single oncogenic pathway characteristic for the differentiation stage of transformed B cells. Mutations not aligned with the central oncogenic driver activated divergent pathways and subverted transformation. Oncogenic lesions in B-ALL frequently mimic cytokine receptor signaling at the pro-B cell stage (through activation of STAT5)2–4 or the pre-B cell receptor in more mature cells (through activation of ERK)5–8. STAT5- and ERK-activating lesions were frequently found but only co-occurred in ~3% of cases (P=2.2E-16). Single-cell mutation and phosphoprotein analyses revealed the segregation of oncogenic STAT5- or ERK-activation to competing clones. STAT5 and ERK engaged opposing biochemical and transcriptional programs orchestrated by MYC and BCL6, respectively. Genetic reactivation of the divergent (suppressed) pathway came at the expense of the principal oncogenic driver and reversed transformation. Conversely, deletion of divergent pathway components accelerated leukemogenesis. Thus, persistence of divergent signaling pathways represents a powerful barrier to transformation while convergence on one principal driver defines a central event in leukemia-initiation. Pharmacological reactivation of suppressed divergent circuits strongly synergized with inhibition of the principal oncogenic driver. Hence, reactivation of divergent pathways can be leveraged as a previously unrecognized strategy to deepen treatment responses.
Collapse
|
33
|
Avoiding or Co-Opting ATP Inhibition: Overview of Type III, IV, V, and VI Kinase Inhibitors. NEXT GENERATION KINASE INHIBITORS 2020. [PMCID: PMC7359047 DOI: 10.1007/978-3-030-48283-1_3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
As described in the previous chapter, most kinase inhibitors that have been developed for use in the clinic act by blocking ATP binding; however, there is growing interest in identifying compounds that target kinase activities and functions without interfering with the conserved features of the ATP-binding site. This chapter will highlight alternative approaches that exploit unique kinase structural features that are being targeted to identify more selective and potent inhibitors. The figure below, adapted from (Sammons et al., Molecular Carcinogenesis 58:1551–1570, 2019), provides a graphical description of the various approaches to manipulate kinase activity. In addition to the type I and II inhibitors, type III kinase inhibitors have been identified to target sites adjacent to the ATP-binding site in the catalytic domain. New information on kinase structure and substrate-binding sites has enabled the identification of type IV kinase inhibitor compounds that target regions outside the catalytic domain. The combination of targeting unique allosteric sites outside the catalytic domain with ATP-targeted compounds has yielded a number of novel bivalent type V kinase inhibitors. Finally, emerging interest in the development of irreversible compounds that form selective covalent interactions with key amino acids involved in kinase functions comprise the class of type VI kinase inhibitors.
Collapse
|
34
|
Elrashedy AA, Ramharack P, Soliman MES. The Perplexity of Synergistic Duality: Inter-molecular Mechanisms of Communication in BCR-ABL1. Anticancer Agents Med Chem 2020; 19:1642-1650. [PMID: 31250767 DOI: 10.2174/1871520619666190620120144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/10/2019] [Accepted: 05/15/2019] [Indexed: 01/23/2023]
Abstract
BACKGROUND Aberrant and proliferative expression of the oncogene BCR-ABL in bone marrow cells is one of the prime causes of Chronic Myeloid Leukemia (CML). It has been established that the tyrosine kinase domain of the BCR-ABL protein is a potential therapeutic target for the treatment of CML. Although the first and second line inhibitors against the enzyme are available, recent studies have indicated that monotherapeutic resistance has become a great challenge. OBJECTIVE In recent studies, the dual inhibition of BCR-ABL by Nilotinib and Asciminib has been shown to overcome drug resistance. This prompted us to investigate the dynamics behind this novel drug combination. METHODS By the utilization of a wide range of computational tools, we defined and compared BCR-ABL's structural and dynamic characteristics when bound as a dual inhibitor system. RESULTS Conformational ensemble analysis presented a sustained inactive protein, as the activation loop, inclusive of the characteristic Tyr257, remained in an open position due to the unassailable binding of Asciminib at the allosteric site. Nilotinib also indicated stronger binding at the catalytic site in the presence of Asciminib, thus exposing new avenues in treating Nilotinib-resistance. This was in accordance with intermolecular hydrogen bond interactions with key binding site residues GLU399, Asn259 and Thr252. CONCLUSION The investigations carried out in this study gave rise to new possibilities in the treatment of resistance in CML, as well as assisting in the design of novel and selective inhibitors as dual anti-cancer drugs.
Collapse
Affiliation(s)
- Ahmed A Elrashedy
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa
| | - Pritika Ramharack
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa
| | - Mahmoud E S Soliman
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa
| |
Collapse
|
35
|
Rizzo AN, Belvitch P, Demeritte R, Garcia JGN, Letsiou E, Dudek SM. Arg mediates LPS-induced disruption of the pulmonary endothelial barrier. Vascul Pharmacol 2020; 128-129:106677. [PMID: 32240815 DOI: 10.1016/j.vph.2020.106677] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/23/2020] [Accepted: 03/21/2020] [Indexed: 12/12/2022]
Abstract
Acute Respiratory Distress Syndrome (ARDS) is a devastating disease process that involves dysregulated inflammation and decreased alveolar-capillary barrier function. Despite increased understanding of the pathophysiology, no effective targeted therapies exist to treat ARDS. Recent preclinical studies suggest that the multi-tyrosine kinase inhibitor, imatinib, which targets the Abl kinases c-Abl and Arg, has the potential to restore endothelial dysfunction caused by inflammatory agonists. Prior work demonstrates that imatinib attenuates LPS (lipopolysaccharide)-induced vascular leak and inflammation; however, the mechanisms underlying these effects remain incompletely understood. In the current study, we demonstrate that imatinib inhibits LPS-induced increase in the phosphorylation of CrkL, a specific substrate of Abl kinases, in human pulmonary endothelial cells. Specific silencing of Arg, and not c-Abl, attenuated LPS-induced pulmonary vascular permeability as measured by electrical cellular impedance sensing (ECIS) and gap formation assays. In addition, direct activation of Abl family kinases with the small molecule activator DPH resulted in endothelial barrier disruption that was attenuated by Arg siRNA. In complementary studies to characterize the mechanisms by which Arg mediates endothelial barrier function, Arg silencing was found to inhibit LPS-induced disruption of adherens junctions and phosphorylation of myosin light chains (MLC). Overall, these results characterize the mechanisms by which imatinib protects against LPS-induced endothelial barrier disruption and suggest that Arg inhibition may represent a novel strategy to enhance endothelial barrier function.
Collapse
Affiliation(s)
- Alicia N Rizzo
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Patrick Belvitch
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Regaina Demeritte
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Joe G N Garcia
- Department of Medicine, University of Arizona, Tucson, AZ, United States of America
| | - Eleftheria Letsiou
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Steven M Dudek
- Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, IL, United States of America.
| |
Collapse
|
36
|
Ferreira Pimentel LC, Cunha AC, Boas Hoelz LV, Canzian HF, Leite Firmino Marinho DI, Boechat N, Bastos MM. Phenylamino-pyrimidine (PAP) Privileged Structure: Synthesis and Medicinal Applications. Curr Top Med Chem 2020; 20:227-243. [DOI: 10.2174/1568026620666200124094949] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/17/2019] [Accepted: 12/25/2019] [Indexed: 12/23/2022]
Abstract
The phenylamino-pyrimidine (PAP) nucleus has been demonstrated to be useful for the development of new drugs and is present in a wide variety of antiretroviral agents and tyrosine kinase inhibitors (TKIs). This review aims to evaluate the application of PAP derivatives in drugs and other bioactive compounds. It was concluded that PAP derivatives are still worth exploring, as they may provide highly competitive ATP TKI’s with nano/picomolar activity.
Collapse
Affiliation(s)
- Luiz Claudio Ferreira Pimentel
- Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos - Fiocruz, Laboratório de Sintese de Farmacos - LASFAR, Manguinhos, CEP 21041-250, Rio de Janeiro, RJ, Brazil
| | - Anna Claudia Cunha
- Universidade Federal Fluminense, Departamento de Quimica Organica, Campus do Valonguinho, CEP 24020-150, Niteroi, RJ, Brazil
| | - Lucas Villas Boas Hoelz
- Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos - Fiocruz, Laboratório de Sintese de Farmacos - LASFAR, Manguinhos, CEP 21041-250, Rio de Janeiro, RJ, Brazil
| | - Henayle Fernandes Canzian
- Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos - Fiocruz, Laboratório de Sintese de Farmacos - LASFAR, Manguinhos, CEP 21041-250, Rio de Janeiro, RJ, Brazil
| | - Debora Inacio Leite Firmino Marinho
- Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos - Fiocruz, Laboratório de Sintese de Farmacos - LASFAR, Manguinhos, CEP 21041-250, Rio de Janeiro, RJ, Brazil
| | - Nubia Boechat
- Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos - Fiocruz, Laboratório de Sintese de Farmacos - LASFAR, Manguinhos, CEP 21041-250, Rio de Janeiro, RJ, Brazil
| | - Monica Macedo Bastos
- Fundacao Oswaldo Cruz, Instituto de Tecnologia em Farmacos, Farmanguinhos - Fiocruz, Laboratório de Sintese de Farmacos - LASFAR, Manguinhos, CEP 21041-250, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
37
|
Paul F, Meng Y, Roux B. Identification of Druggable Kinase Target Conformations Using Markov Model Metastable States Analysis of apo-Abl. J Chem Theory Comput 2020; 16:1896-1912. [PMID: 31999924 DOI: 10.1021/acs.jctc.9b01158] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Kinases are important targets for drug development. However, accounting for the impact of possible structural rearrangements on the binding of kinase inhibitors is complicated by the extensive flexibility of their catalytic domain. The dynamic N-lobe contains four particular mobile structural elements: the Asp-Phe-Gly (DFG) motif, the phosphate (P) positioning loop, the activation (A) loop, and the αC helix. In our previous study [Meng et al. J. Chem. Theory Comput. 2018 14, 2721-2732], we combined various simulation techniques with Markov state modeling (MSM) to explore the free energy landscape of Abl kinase beyond conformations that are known from X-ray crystallography. Here we examine the resulting Markov model in greater detail by analyzing its metastable states. A characterization of the states in terms of their DFG state, P-loop, and αC conformations is presented and compared to existing classification schemes. Several metastable states are found to be structurally close to known crystal structures of different kinases in complex with a variety of inhibitors. These results suggest that the set of conformations accessible to tyrosine kinases may be shared within the entire family and that the conformational dynamics of one kinase in the absence of any ligand can provide meaningful information about possible target conformations for inhibitors of any member of the kinase family.
Collapse
Affiliation(s)
- Fabian Paul
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois 60637-1454, United States
| | - Yilin Meng
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois 60637-1454, United States
| | - Benoît Roux
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois 60637-1454, United States
| |
Collapse
|
38
|
Fogha J, Diharce J, Obled A, Aci-Sèche S, Bonnet P. Computational Analysis of Crystallization Additives for the Identification of New Allosteric Sites. ACS OMEGA 2020; 5:2114-2122. [PMID: 32064372 PMCID: PMC7016913 DOI: 10.1021/acsomega.9b02697] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/21/2019] [Indexed: 06/10/2023]
Abstract
Allosteric effect can modulate the biological activity of a protein. Thus, the discovery of new allosteric sites is very attractive for designing new modulators or inhibitors. Here, we propose an innovative way to identify allosteric sites, based on crystallization additives (CA), used to stabilize proteins during the crystallization process. Density and clustering analyses of CA, applied on protein kinase and nuclear receptor families, revealed that CA are not randomly distributed around protein structures, but they tend to aggregate near common sites. All orthosteric and allosteric cavities described in the literature are retrieved from the analysis of CA distribution. In addition, new sites were identified, which could be associated to putative allosteric sites. We proposed an efficient and easy way to use the structural information of CA to identify allosteric sites. This method could assist medicinal chemists for the design of new allosteric compounds targeting cavities of new drug targets.
Collapse
|
39
|
Hoshi Y, Uchida Y, Tachikawa M, Ohtsuki S, Couraud PO, Suzuki T, Terasaki T. Oxidative stress-induced activation of Abl and Src kinases rapidly induces P-glycoprotein internalization via phosphorylation of caveolin-1 on tyrosine-14, decreasing cortisol efflux at the blood-brain barrier. J Cereb Blood Flow Metab 2020; 40:420-436. [PMID: 30621530 PMCID: PMC7370610 DOI: 10.1177/0271678x18822801] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Exposure of the brain to high levels of glucocorticoids during ischemia-reperfusion induces neuronal cell death. Oxidative stress alters blood-brain barrier (BBB) function during ischemia-reperfusion, and so we hypothesized that it might impair P-glycoprotein (P-gp)-mediated efflux transport of glucocorticoids at the BBB. Therefore, the purpose of this study was to clarify the molecular mechanism of this putative decrease of P-gp-mediated efflux function. First, we established that H2O2 treatment of a human in vitro BBB model (hCMEC/D3) reduced both P-gp efflux transport activity and protein expression on the plasma membrane within 20 min. These results suggested that the rapid decrease of efflux function might be due to internalization of P-gp. Furthermore, H2O2 treatment markedly increased tyrosine-14-phosphorylated caveolin-1, which is involved in P-gp internalization. A brain perfusion study in rats showed that cortisol efflux at the BBB was markedly decreased by H2O2 administration, and inhibitors of Abl kinase and Src kinase, which phosphorylate tyrosine-14 in caveolin-1, suppressed this decrease. Overall, these findings support the idea that oxidative stress-induced activation of Abl kinase and Src kinase induces internalization of P-gp via the phosphorylation of tyrosine-14 in caveolin-1, leading to a rapid decrease of P-gp-mediated cortisol efflux at the BBB.
Collapse
Affiliation(s)
- Yutaro Hoshi
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yasuo Uchida
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Masanori Tachikawa
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Sumio Ohtsuki
- Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | | | - Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tetsuya Terasaki
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
40
|
Abstract
Pseudokinases are members of the protein kinase superfamily but signal primarily through noncatalytic mechanisms. Many pseudokinases contribute to the pathologies of human diseases, yet they remain largely unexplored as drug targets owing to challenges associated with modulation of their biological functions. Our understanding of the structure and physiological roles of pseudokinases has improved substantially over the past decade, revealing intriguing similarities between pseudokinases and their catalytically active counterparts. Pseudokinases often adopt conformations that are analogous to those seen in catalytically active kinases and, in some cases, can also bind metal cations and/or nucleotides. Several clinically approved kinase inhibitors have been shown to influence the noncatalytic functions of active kinases, providing hope that similar properties in pseudokinases could be pharmacologically regulated. In this Review, we discuss known roles of pseudokinases in disease, their unique structural features and the progress that has been made towards developing pseudokinase-directed therapeutics.
Collapse
|
41
|
Wang H, Guan Z, Qiu J, Jia Y, Zeng C, Zhao Y. Novel method to identify group-specific non-catalytic pockets of human kinome for drug design. RSC Adv 2020; 10:2004-2015. [PMID: 35494619 PMCID: PMC9047066 DOI: 10.1039/c9ra07471f] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 12/27/2019] [Indexed: 01/11/2023] Open
Abstract
Kinase proteins have been intensively investigated as drug targets for decades because of their crucial involvement in many biological pathways. We developed hybrid approach to identify non-catalytic pockets and will benefit the kinome drug design.
Collapse
Affiliation(s)
- Huiwen Wang
- Department of Physics
- Institute of Biophysics
- Central China Normal University
- Wuhan 430079
- China
| | - Zeyu Guan
- Department of Physics
- Institute of Biophysics
- Central China Normal University
- Wuhan 430079
- China
| | - Jiadi Qiu
- Department of Physics
- Institute of Biophysics
- Central China Normal University
- Wuhan 430079
- China
| | - Ya Jia
- Department of Physics
- Institute of Biophysics
- Central China Normal University
- Wuhan 430079
- China
| | - Chen Zeng
- Department of Physics
- Institute of Biophysics
- Central China Normal University
- Wuhan 430079
- China
| | - Yunjie Zhao
- Department of Physics
- Institute of Biophysics
- Central China Normal University
- Wuhan 430079
- China
| |
Collapse
|
42
|
Essential site scanning analysis: A new approach for detecting sites that modulate the dispersion of protein global motions. Comput Struct Biotechnol J 2020; 18:1577-1586. [PMID: 32637054 PMCID: PMC7330491 DOI: 10.1016/j.csbj.2020.06.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022] Open
Abstract
Despite the wealth of methods developed for exploring the molecular basis of allostery in biomolecular systems, there is still a need for structure-based predictive tools that can efficiently detect susceptible sites for triggering allosteric responses. Toward this goal, we introduce here an elastic network model (ENM)-based method, Essential Site Scanning Analysis (ESSA). Essential sites are here defined as residues that would significantly alter the protein's global dynamics if bound to a ligand. To mimic the crowding induced upon substrate binding, the heavy atoms of each residue are incorporated as additional network nodes into the α-carbon-based ENM, and the resulting shifts in soft mode frequencies are used as a metric for evaluating the essentiality of each residue. Results on a dataset of monomeric proteins indicate the enrichment of allosteric and orthosteric binding sites, as well as global hinge regions among essential residues, highlighting the significant role of these sites in controlling the overall structural dynamics. Further integration of ESSA with information on predicted pockets and their local hydrophobicity density enables successful predictions of allosteric pockets for both ligand-bound and -unbound structures. ESSA can be efficiently applied to large multimeric systems. Three case studies, namely (i) G-protein binding to a GPCR, (ii) heterotrimeric assembly of the Ser/Thr protein phosphatase PP2A, and (iii) allo-targeting of AMPA receptor, demonstrate the utility of ESSA for identifying essential sites and narrowing down target allosteric sites identified by druggability simulations.
Collapse
|
43
|
Kinase-independent inhibition of cyclophosphamide-induced pathways protects the ovarian reserve and prolongs fertility. Cell Death Dis 2019; 10:726. [PMID: 31562295 PMCID: PMC6765024 DOI: 10.1038/s41419-019-1961-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 08/07/2019] [Accepted: 08/27/2019] [Indexed: 01/27/2023]
Abstract
Premature ovarian failure and infertility are adverse effects of cancer therapies. The mechanism underlying chemotherapy-mediated depletion of the ovarian reserve remains unclear. Here, we aim to identify the signaling pathways involved in the loss of the ovarian reserve to prevent the damaging effects of chemotherapy. We evaluated the effects of cyclophosphamide, one of the most damaging chemotherapeutic drugs, against follicle reserve. In vivo studies showed that the cyclophosphamide-induced loss of ovarian reserve occurred through a sequential mechanism. Cyclophosphamide exposure induced the activation of both DNAPK-γH2AX-checkpoint kinase 2 (CHK2)-p53/TAp63α isoform and protein kinase B (AKT)-forkhead box O3 (FOXO3a) signaling axes in the nucleus of oocytes. Concomitant administration of an allosteric ABL inhibitor and cyclophosphamide modulated both pathways while protecting the ovarian reserve from chemotherapy assaults. As a consequence, the fertility of the treated mice was prolonged. On the contrary, the administration of an allosteric ABL activator enhanced the lethal effects of cyclophosphamide while shortening mouse fertility. Therefore, kinase-independent inhibition may serve as an effective ovarian-protective strategy in women under chemotherapy.
Collapse
|
44
|
Astl L, Verkhivker GM. Data-driven computational analysis of allosteric proteins by exploring protein dynamics, residue coevolution and residue interaction networks. Biochim Biophys Acta Gen Subj 2019:S0304-4165(19)30179-5. [PMID: 31330173 DOI: 10.1016/j.bbagen.2019.07.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 07/15/2019] [Accepted: 07/17/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Computational studies of allosteric interactions have witnessed a recent renaissance fueled by the growing interest in modeling of the complex molecular assemblies and biological networks. Allosteric interactions in protein structures allow for molecular communication in signal transduction networks. METHODS In this work, we performed a large scale comprehensive and multi-faceted analysis of >300 diverse allosteric proteins and complexes with allosteric modulators. By modeling and exploring coarse-grained dynamics, residue coevolution, and residue interaction networks for allosteric proteins, we have determined unifying molecular signatures shared by allosteric systems. RESULTS The results of this study have suggested that allosteric inhibitors and allosteric activators may differentially affect global dynamics and network organization of protein systems, leading to diverse allosteric mechanisms. By using structural and functional data on protein kinases, we present a detailed case study that that included atomic-level analysis of coevolutionary networks in kinases bound with allosteric inhibitors and activators. CONCLUSIONS We have found that coevolutionary networks can form direct communication pathways connecting functional regions and can recapitulate key regulatory sites and interactions responsible for allosteric signaling in the studied protein systems. The results of this computational investigation are compared with the experimental studies and reveal molecular signatures of known regulatory hotspots in protein kinases. GENERAL SIGNIFICANCE This study has shown that allosteric inhibitors and allosteric activators can have a different effect on residue interaction networks and can exploit distinct regulatory mechanisms, which could open up opportunities for probing allostery and new drug combinations with broad range of activities.
Collapse
Affiliation(s)
- Lindy Astl
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, United States of America
| | - Gennady M Verkhivker
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, United States of America; Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, United States of America.
| |
Collapse
|
45
|
Error Tolerance of Machine Learning Algorithms across Contemporary Biological Targets. Molecules 2019; 24:molecules24112115. [PMID: 31167452 PMCID: PMC6601015 DOI: 10.3390/molecules24112115] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/31/2019] [Accepted: 06/01/2019] [Indexed: 12/16/2022] Open
Abstract
Machine learning continues to make strident advances in the prediction of desired properties concerning drug development. Problematically, the efficacy of machine learning in these arenas is reliant upon highly accurate and abundant data. These two limitations, high accuracy and abundance, are often taken together; however, insight into the dataset accuracy limitation of contemporary machine learning algorithms may yield insight into whether non-bench experimental sources of data may be used to generate useful machine learning models where there is a paucity of experimental data. We took highly accurate data across six kinase types, one GPCR, one polymerase, a human protease, and HIV protease, and intentionally introduced error at varying population proportions in the datasets for each target. With the generated error in the data, we explored how the retrospective accuracy of a Naïve Bayes Network, a Random Forest Model, and a Probabilistic Neural Network model decayed as a function of error. Additionally, we explored the ability of a training dataset with an error profile resembling that produced by the Free Energy Perturbation method (FEP+) to generate machine learning models with useful retrospective capabilities. The categorical error tolerance was quite high for a Naïve Bayes Network algorithm averaging 39% error in the training set required to lose predictivity on the test set. Additionally, a Random Forest tolerated a significant degree of categorical error introduced into the training set with an average error of 29% required to lose predictivity. However, we found the Probabilistic Neural Network algorithm did not tolerate as much categorical error requiring an average of 20% error to lose predictivity. Finally, we found that a Naïve Bayes Network and a Random Forest could both use datasets with an error profile resembling that of FEP+. This work demonstrates that computational methods of known error distribution like FEP+ may be useful in generating machine learning models not based on extensive and expensive in vitro-generated datasets.
Collapse
|
46
|
Astl L, Verkhivker GM. Atomistic Modeling of the ABL Kinase Regulation by Allosteric Modulators Using Structural Perturbation Analysis and Community-Based Network Reconstruction of Allosteric Communications. J Chem Theory Comput 2019; 15:3362-3380. [PMID: 31017783 DOI: 10.1021/acs.jctc.9b00119] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In this work, we have examined the molecular mechanisms of allosteric regulation of the ABL tyrosine kinase at the atomic level. Atomistic modeling of the ABL complexes with a panel of allosteric modulators has been performed using a combination of molecular dynamics simulations, structural residue perturbation scanning, and a novel community analysis of the residue interaction networks. Our results have indicated that allosteric inhibitors and activators may exert a differential control on allosteric signaling between the kinase binding sites and functional regions. While the inhibitor binding can strengthen the closed ABL state and induce allosteric communications directed from the allosteric pocket to the ATP binding site, the DPH activator may induce a more dynamic open form and activate allosteric couplings between the ATP and substrate binding sites. By leveraging a network-centric theoretical framework, we have introduced a novel community analysis method and global topological parameters that have unveiled the hierarchical modularity and the intercommunity bridging sites in the residue interaction network. We have found that allosteric functional hotspots responsible for the kinase regulation may serve the intermodular bridges in the global interaction network. The central conclusion from this analysis is that the regulatory switch centers play a fundamental role in the modular network organization of ABL as the unique intercommunity bridges that connect the SH2 and SH3 domains with the catalytic core into a functional kinase assembly. The hierarchy of network organization in the ABL regulatory complexes may allow for the synergistic action of dense intercommunity links required for the robust signal transfer in the catalytic core and sparse network bridges acting as the regulatory control points that orchestrate allosteric transitions between the inhibited and active kinase forms.
Collapse
Affiliation(s)
- Lindy Astl
- Graduate Program in Computational and Data Sciences, Keck Center for Science and Engineering, Schmid College of Science and Technology , Chapman University , One University Drive , Orange , California 92866 , United States
| | - Gennady M Verkhivker
- Graduate Program in Computational and Data Sciences, Keck Center for Science and Engineering, Schmid College of Science and Technology , Chapman University , One University Drive , Orange , California 92866 , United States.,Department of Biomedical and Pharmaceutical Sciences , Chapman University School of Pharmacy , Irvine , California 92618 , United States
| |
Collapse
|
47
|
Olabarria M, Pasini S, Corona C, Robador P, Song C, Patel H, Lefort R. Dysfunction of the ubiquitin ligase E3A Ube3A/E6-AP contributes to synaptic pathology in Alzheimer's disease. Commun Biol 2019; 2:111. [PMID: 30937395 PMCID: PMC6430817 DOI: 10.1038/s42003-019-0350-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 01/23/2019] [Indexed: 12/21/2022] Open
Abstract
Synaptic dysfunction and synapse loss are prominent features in Alzheimer's disease. Members of the Rho-family of guanosine triphosphatases, specifically RhoA, and the synaptic protein Arc are implicated in these pathogenic processes. They share a common regulatory molecule, the E3 ligase Ube3A/E6-AP. Here, we show that Ube3A is reduced in an Alzheimer's disease mouse model, Tg2576 mouse, which overexpresses human APP695 carrying the Swedish mutation, and accumulates Aβ in the brain. Depletion of Ube3A precedes the age-dependent behavioral deficits and loss of dendritic spines in these mice, and results from a decrease in solubility following phosphorylation by c-Abl, after Aβ exposure. Loss of Ube3A triggers the accumulation of Arc and Ephexin-5, driving internalization of GluR1, and activation of RhoA, respectively, culminating in pruning of synapses, which is blocked by restoring Ube3A. Taken together, our results place Ube3A as a critical player in Alzheimer's disease pathogenesis, and as a potential therapeutic target.
Collapse
Affiliation(s)
- Markel Olabarria
- Taub Institute for Research on Alzheimer’s Disease & the Aging Brain and the Department of Pathology & Cell Biology, Columbia University, New York, NY 10032 USA
| | - Silvia Pasini
- Taub Institute for Research on Alzheimer’s Disease & the Aging Brain and the Department of Pathology & Cell Biology, Columbia University, New York, NY 10032 USA
- Present Address: Department of Ophthalmology and Visual Sciences, Vanderbilt University Medical Center, Nashville, TN 37205 USA
| | - Carlo Corona
- Taub Institute for Research on Alzheimer’s Disease & the Aging Brain and the Department of Pathology & Cell Biology, Columbia University, New York, NY 10032 USA
| | - Pablo Robador
- Taub Institute for Research on Alzheimer’s Disease & the Aging Brain and the Department of Pathology & Cell Biology, Columbia University, New York, NY 10032 USA
| | - Cheng Song
- Taub Institute for Research on Alzheimer’s Disease & the Aging Brain and the Department of Pathology & Cell Biology, Columbia University, New York, NY 10032 USA
| | - Hardik Patel
- Taub Institute for Research on Alzheimer’s Disease & the Aging Brain and the Department of Pathology & Cell Biology, Columbia University, New York, NY 10032 USA
| | - Roger Lefort
- Taub Institute for Research on Alzheimer’s Disease & the Aging Brain and the Department of Pathology & Cell Biology, Columbia University, New York, NY 10032 USA
| |
Collapse
|
48
|
Simpson GL, Bertrand SM, Borthwick JA, Campobasso N, Chabanet J, Chen S, Coggins J, Cottom J, Christensen SB, Dawson HC, Evans HL, Hobbs AN, Hong X, Mangatt B, Munoz-Muriedas J, Oliff A, Qin D, Scott-Stevens P, Ward P, Washio Y, Yang J, Young RJ. Identification and Optimization of Novel Small c-Abl Kinase Activators Using Fragment and HTS Methodologies. J Med Chem 2019; 62:2154-2171. [PMID: 30689376 DOI: 10.1021/acs.jmedchem.8b01872] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Abelson kinase (c-Abl) is a ubiquitously expressed, nonreceptor tyrosine kinase which plays a key role in cell differentiation and survival. It was hypothesized that transient activation of c-Abl kinase via displacement of the N-terminal autoinhibitory "myristoyl latch", may lead to an increased hematopoietic stem cell differentiation. This would increase the numbers of circulating neutrophils and so be an effective treatment for chemotherapy-induced neutropenia. This paper describes the discovery and optimization of a thiazole series of novel small molecule c-Abl activators, initially identified by a high throughput screening. Subsequently, a scaffold-hop, which exploited the improved physicochemical properties of a dihydropyrazole analogue, identified through fragment screening, delivered potent, soluble, cell-active c-Abl activators, which demonstrated the intracellular activation of c-Abl in vivo.
Collapse
Affiliation(s)
- Graham L Simpson
- Medicines Research Centre , GlaxoSmithKline R&D , Gunnels Wood Road , Stevenage , Hertfordshire SG1 2NY , U.K
| | - Sophie M Bertrand
- Medicines Research Centre , GlaxoSmithKline R&D , Gunnels Wood Road , Stevenage , Hertfordshire SG1 2NY , U.K
| | - Jennifer A Borthwick
- Medicines Research Centre , GlaxoSmithKline R&D , Gunnels Wood Road , Stevenage , Hertfordshire SG1 2NY , U.K
| | - Nino Campobasso
- GlaxoSmithKline R&D , 1250 South Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Julien Chabanet
- Medicines Research Centre , GlaxoSmithKline R&D , Gunnels Wood Road , Stevenage , Hertfordshire SG1 2NY , U.K
| | | | - Julia Coggins
- Medicines Research Centre , GlaxoSmithKline R&D , Gunnels Wood Road , Stevenage , Hertfordshire SG1 2NY , U.K
| | - Josh Cottom
- GlaxoSmithKline R&D , 1250 South Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | | | - Helen C Dawson
- Medicines Research Centre , GlaxoSmithKline R&D , Gunnels Wood Road , Stevenage , Hertfordshire SG1 2NY , U.K
| | - Helen L Evans
- Medicines Research Centre , GlaxoSmithKline R&D , Gunnels Wood Road , Stevenage , Hertfordshire SG1 2NY , U.K
| | - Andrew N Hobbs
- Medicines Research Centre , GlaxoSmithKline R&D , Gunnels Wood Road , Stevenage , Hertfordshire SG1 2NY , U.K
| | - Xuan Hong
- GlaxoSmithKline R&D , 1250 South Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Biju Mangatt
- GlaxoSmithKline R&D , 1250 South Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Jordi Munoz-Muriedas
- Medicines Research Centre , GlaxoSmithKline R&D , Gunnels Wood Road , Stevenage , Hertfordshire SG1 2NY , U.K
| | - Allen Oliff
- GlaxoSmithKline R&D , 1250 South Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Donghui Qin
- Medicines Research Centre , GlaxoSmithKline R&D , Gunnels Wood Road , Stevenage , Hertfordshire SG1 2NY , U.K
| | - Paul Scott-Stevens
- Medicines Research Centre , GlaxoSmithKline R&D , Gunnels Wood Road , Stevenage , Hertfordshire SG1 2NY , U.K
| | - Paris Ward
- GlaxoSmithKline R&D , 1250 South Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Yoshiaki Washio
- Medicines Research Centre , GlaxoSmithKline R&D , Gunnels Wood Road , Stevenage , Hertfordshire SG1 2NY , U.K
| | - Jingsong Yang
- GlaxoSmithKline R&D , 1250 South Collegeville Road , Collegeville , Pennsylvania 19426 , United States
| | - Robert J Young
- Medicines Research Centre , GlaxoSmithKline R&D , Gunnels Wood Road , Stevenage , Hertfordshire SG1 2NY , U.K
| |
Collapse
|
49
|
Astl L, Tse A, Verkhivker GM. Interrogating Regulatory Mechanisms in Signaling Proteins by Allosteric Inhibitors and Activators: A Dynamic View Through the Lens of Residue Interaction Networks. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1163:187-223. [DOI: 10.1007/978-981-13-8719-7_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
50
|
Schoepfer J, Jahnke W, Berellini G, Buonamici S, Cotesta S, Cowan-Jacob SW, Dodd S, Drueckes P, Fabbro D, Gabriel T, Groell JM, Grotzfeld RM, Hassan AQ, Henry C, Iyer V, Jones D, Lombardo F, Loo A, Manley PW, Pellé X, Rummel G, Salem B, Warmuth M, Wylie AA, Zoller T, Marzinzik AL, Furet P. Discovery of Asciminib (ABL001), an Allosteric Inhibitor of the Tyrosine Kinase Activity of BCR-ABL1. J Med Chem 2018; 61:8120-8135. [DOI: 10.1021/acs.jmedchem.8b01040] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Joseph Schoepfer
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Wolfgang Jahnke
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | | | | | - Simona Cotesta
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Sandra W. Cowan-Jacob
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Stephanie Dodd
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Peter Drueckes
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | | | - Tobias Gabriel
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Jean-Marc Groell
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Robert M. Grotzfeld
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | | | - Chrystèle Henry
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | | | - Darryl Jones
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | | | - Alice Loo
- Novartis Institutes for BioMedical Research, 250 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Paul W. Manley
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Xavier Pellé
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Gabriele Rummel
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Bahaa Salem
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | | | | | - Thomas Zoller
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Andreas L. Marzinzik
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Pascal Furet
- Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| |
Collapse
|