1
|
Jin S, Zhang M, Qiao X. Cyclophilin A: promising target in cancer therapy. Cancer Biol Ther 2024; 25:2425127. [PMID: 39513594 PMCID: PMC11552246 DOI: 10.1080/15384047.2024.2425127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/08/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
Cyclophilin A (CypA), a member of the immunophilin family, stands out as the most prevalent among the cyclophilins found in humans. Beyond serving as the intracellular receptor for the immunosuppressive drug cyclosporine A (CsA), CypA exerts critical functions within the cell via its peptidyl-prolyl cis-trans isomerase (PPIase) activity, which is crucial for processes, such as protein folding, trafficking, assembly, modulation of immune responses, and cell signaling. Increasing evidence indicates that CypA is up-regulated in a variety of human cancers and it may be a novel potential therapeutic target for cancer treatment. Therefore, gaining a thorough understanding of CypA's contribution to cancer could yield fresh perspectives and inform the development of innovative therapeutic approaches. This review delves into the multifaceted roles of CypA in cancer biology and explores the therapeutic potential of targeting CypA.
Collapse
Affiliation(s)
- Shujuan Jin
- Shenzhen Institute for Technology Innovation, National Institute of Metrology, Shenzhen, Guangdong, China
| | - Mengjiao Zhang
- Chenxi Women’s and Children’s Hospital, Huaihua, Hunan, China
| | - Xiaoting Qiao
- Shenzhen Institute for Technology Innovation, National Institute of Metrology, Shenzhen, Guangdong, China
| |
Collapse
|
2
|
Kage M, Hayashi R, Matsuo A, Tamiya M, Kuramoto S, Ohara K, Irie M, Chiyoda A, Takano K, Ito T, Kotake T, Takeyama R, Ishikawa S, Nomura K, Furuichi N, Morita Y, Hashimoto S, Kawada H, Nishimura Y, Nii K, Sase H, Ohta A, Kojima T, Iikura H, Tanada M, Shiraishi T. Structure-activity relationships of middle-size cyclic peptides, KRAS inhibitors derived from an mRNA display. Bioorg Med Chem 2024; 110:117830. [PMID: 38981216 DOI: 10.1016/j.bmc.2024.117830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/11/2024]
Abstract
Cyclic peptides are attracting attention as therapeutic agents due to their potential for oral absorption and easy access to tough intracellular targets. LUNA18, a clinical KRAS inhibitor, was transformed-without scaffold hopping-from the initial hit by using an mRNA display library that met our criteria for drug-likeness. In drug discovery using mRNA display libraries, hit compounds always possess a site linked to an mRNA tag. Here, we describe our examination of the Structure-Activity Relationship (SAR) using X-ray structures for chemical optimization near the site linked to the mRNA tag, equivalent to the C-terminus. Structural modifications near the C-terminus demonstrated a relatively wide range of tolerance for side chains. Furthermore, we show that a single atom modification is enough to change the pharmacokinetic (PK) profile. Since there are four positions where side chain modification is permissible in terms of activity, it is possible to flexibly adjust the pharmacokinetic profile by structurally optimizing the side chain. The side chain transformation findings demonstrated here may be generally applicable to hits obtained from mRNA display libraries.
Collapse
Affiliation(s)
- Mirai Kage
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Ryuji Hayashi
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan.
| | - Atsushi Matsuo
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Minoru Tamiya
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Shino Kuramoto
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Kazuhiro Ohara
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Machiko Irie
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Aya Chiyoda
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Koji Takano
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Toshiya Ito
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Tomoya Kotake
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Ryuuichi Takeyama
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Shiho Ishikawa
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Kenichi Nomura
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Noriyuki Furuichi
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Yuya Morita
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Satoshi Hashimoto
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Hatsuo Kawada
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Yoshikazu Nishimura
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Keiji Nii
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Hitoshi Sase
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Atsushi Ohta
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Tetsuo Kojima
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Hitoshi Iikura
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan
| | - Mikimasa Tanada
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan.
| | - Takuya Shiraishi
- Research Division, Chugai Pharmaceutical Co. Ltd., 216, Totsuka-cho, Totsuka-ku, Yokohama, Kanagawa 244-8602, Japan.
| |
Collapse
|
3
|
Flaxman HA, Chrysovergi MA, Han H, Kabir F, Lister RT, Chang CF, Yvon R, Black KE, Weigert A, Savai R, Egea-Zorrilla A, Pardo-Saganta A, Lagares D, Woo CM. Sanglifehrin A mitigates multiorgan fibrosis by targeting the collagen chaperone cyclophilin B. JCI Insight 2024; 9:e171162. [PMID: 38900587 PMCID: PMC11383833 DOI: 10.1172/jci.insight.171162] [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: 04/04/2023] [Accepted: 06/18/2024] [Indexed: 06/22/2024] Open
Abstract
Pathological deposition and crosslinking of collagen type I by activated myofibroblasts drives progressive tissue fibrosis. Therapies that inhibit collagen synthesis have potential as antifibrotic agents. We identify the collagen chaperone cyclophilin B as a major cellular target of the natural product sanglifehrin A (SfA) using photoaffinity labeling and chemical proteomics. Mechanistically, SfA inhibits and induces the secretion of cyclophilin B from the endoplasmic reticulum (ER) and prevents TGF-β1-activated myofibroblasts from synthesizing and secreting collagen type I in vitro, without inducing ER stress or affecting collagen type I mRNA transcription, myofibroblast migration, contractility, or TGF-β1 signaling. In vivo, SfA induced cyclophilin B secretion in preclinical models of fibrosis, thereby inhibiting collagen synthesis from fibrotic fibroblasts and mitigating the development of lung and skin fibrosis in mice. Ex vivo, SfA induces cyclophilin B secretion and inhibits collagen type I secretion from fibrotic human lung fibroblasts and samples from patients with idiopathic pulmonary fibrosis (IPF). Taken together, we provide chemical, molecular, functional, and translational evidence for demonstrating direct antifibrotic activities of SfA in preclinical and human ex vivo fibrotic models. Our results identify the cellular target of SfA, the collagen chaperone cyclophilin B, as a mechanistic target for the treatment of organ fibrosis.
Collapse
Affiliation(s)
- Hope A Flaxman
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Maria-Anna Chrysovergi
- Fibrosis Research Center, Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hongwei Han
- Fibrosis Research Center, Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Farah Kabir
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Rachael T Lister
- Fibrosis Research Center, Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Chia-Fu Chang
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Robert Yvon
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Katharine E Black
- Fibrosis Research Center, Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andreas Weigert
- Goethe-University Frankfurt, Faculty of Medicine, Institute of Biochemistry I, Germany
- Frankfurt Cancer Institute (FCI), Goethe University, and German Cancer Consortium (DKTK), Germany
| | - Rajkumar Savai
- Frankfurt Cancer Institute (FCI), Goethe University, and German Cancer Consortium (DKTK), Germany
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
- Institute for Lung Health (ILH), Department of Internal Medicine, Justus-Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), DZL, Giessen, Germany
- Cardio-Pulmonary Institute (CPI), Department of Internal Medicine, Justus Liebig University, Giessen, Germany
| | - Alejandro Egea-Zorrilla
- Institute for Lung Health (ILH), Department of Internal Medicine, Justus-Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), DZL, Giessen, Germany
- Cardio-Pulmonary Institute (CPI), Department of Internal Medicine, Justus Liebig University, Giessen, Germany
| | - Ana Pardo-Saganta
- Institute for Lung Health (ILH), Department of Internal Medicine, Justus-Liebig University, Universities of Giessen and Marburg Lung Center (UGMLC), DZL, Giessen, Germany
- Cardio-Pulmonary Institute (CPI), Department of Internal Medicine, Justus Liebig University, Giessen, Germany
| | - David Lagares
- Fibrosis Research Center, Center for Immunology and Inflammatory Diseases, Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Christina M Woo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
4
|
Guareschi F, Del Favero E, Ricci C, Cantù L, Brandolini M, Sambri V, Nicoli S, Pescina S, D'Angelo D, Rossi I, Buttini F, Bettini R, Sonvico F. Cyclosporine A micellar nasal spray characterization and antiviral action against SARS-CoV-2. Eur J Pharm Sci 2024; 193:106673. [PMID: 38103657 DOI: 10.1016/j.ejps.2023.106673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023]
Abstract
The upper airways represent the point of entrance from where Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection spreads to the lungs. In the present work, α-tocopheryl-polyethylene-glycol succinate (TPGS) micelles loaded with cyclosporine A (CSA) were developed for nasal administration to prevent or treat the viral infection in the very first phases. The behavior of the micelles in presence of simulated nasal mucus was investigated in terms of stability and mucopenetration rate, evidencing long-term stability and fast diffusion across the glycoproteins matrix. Moreover, the spray characteristics of the micellar formulation and deposition profile in a silicon nasal model were studied using three nasal spray devices. Results allowed to identify the nasal spray pump (BiVax, Aptar) able to provide the wider and uniform deposition of the nasal cavity. The cyclosporine A micelles antiviral activity against SARS-CoV-2 was tested on the Omicron BA.1 variant using Vero E6 cells with protocols simulating treatment before, during and after the infection of the upper airways. Complete viral inactivation was observed for the cyclosporine-loaded micelles while a very low activity was evidenced for the non-formulated drug, suggesting a synergistic activity of the drug and the formulation. In conclusion, this work showed that the developed cyclosporine A-loaded micellar formulations have the potential to be clinically effective against a wide spectrum of coronavirus variants.
Collapse
Affiliation(s)
- Fabiola Guareschi
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/a, 43124 Parma, Italy
| | - Elena Del Favero
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Fratelli Cervi 93, 20054 Milan, Italy
| | - Caterina Ricci
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Fratelli Cervi 93, 20054 Milan, Italy
| | - Laura Cantù
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Fratelli Cervi 93, 20054 Milan, Italy
| | - Martina Brandolini
- Unit of Microbiology, The Great Romagna Hub Laboratory, Piazza della Liberazione 60, 47522 Pievesestina, Italy
| | - Vittorio Sambri
- Unit of Microbiology, The Great Romagna Hub Laboratory, Piazza della Liberazione 60, 47522 Pievesestina, Italy; Department of Experimental, Diagnostic and Specialty Medicine-DIMES, Alma Mater Studiorum-University of Bologna, Via Massarenti 1, 40138 Bologna, Italy
| | - Sara Nicoli
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/a, 43124 Parma, Italy
| | - Silvia Pescina
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/a, 43124 Parma, Italy
| | - Davide D'Angelo
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/a, 43124 Parma, Italy
| | - Irene Rossi
- Nanopharm Ltd, Franklin House, Grange Road, Cwmbran NP44 3WY, United Kingdom
| | - Francesca Buttini
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/a, 43124 Parma, Italy; Interdepartmental Center for Innovation in Health Products, Biopharmanet_TEC, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Ruggero Bettini
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/a, 43124 Parma, Italy; Interdepartmental Center for Innovation in Health Products, Biopharmanet_TEC, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy
| | - Fabio Sonvico
- ADDRes Lab, Department of Food and Drug, University of Parma, Parco Area delle Scienze 27/a, 43124 Parma, Italy; Interdepartmental Center for Innovation in Health Products, Biopharmanet_TEC, University of Parma, Parco Area delle Scienze 27/A, 43124 Parma, Italy.
| |
Collapse
|
5
|
Stauffer W, Bobardt M, Ure D, Foster R, Gallay P. The Cyclophilin Inhibitor Rencofilstat Decreases HCV-Induced Hepatocellular Carcinoma Independently of Its Antiviral Activity. Viruses 2023; 15:2099. [PMID: 37896876 PMCID: PMC10612079 DOI: 10.3390/v15102099] [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: 09/01/2023] [Revised: 10/07/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
There is an urgent need for the identification of new drugs that inhibit HCV-induced hepatocellular carcinoma (HCC). Our work demonstrates that cyclophilin inhibitors (CypIs) represent such new drugs. We demonstrate that the nonimmunosuppressive cyclosporine A (CsA) analog (CsAa) rencofilstat possesses dual therapeutic activities for the treatment of HCV infection and HCV-induced HCC. Specifically, we show that the HCV infection of humanized mice results in the progressive development of HCC. This is true for the four genotypes tested (1 to 4). Remarkably, we demonstrate that rencofilstat inhibits the development of HCV-induced HCC in mice even when added 16 weeks after infection when HCC is well established. Importantly, we show that rencofilstat drastically reduces HCC progression independently of its anti-HCV activity. Indeed, the CypI rencofilstat inhibits HCC, while other anti-HCV agents such as NS5A (NS5Ai) and NS5B (NS5Bi) fail to reduce HCC. In conclusion, this study shows for the first time that the CypI rencofilstat represents a potent therapeutic agent for the treatment of HCV-induced HCC.
Collapse
Affiliation(s)
- Winston Stauffer
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; (W.S.); (M.B.)
| | - Michael Bobardt
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; (W.S.); (M.B.)
| | - Daren Ure
- Hepion Pharmaceuticals Inc., Edison, NJ 08837, USA; (D.U.); (R.F.)
| | - Robert Foster
- Hepion Pharmaceuticals Inc., Edison, NJ 08837, USA; (D.U.); (R.F.)
| | - Philippe Gallay
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; (W.S.); (M.B.)
| |
Collapse
|
6
|
Stauffer W, Bobardt M, Ure D, Foster R, Gallay P. The Cyclophilin Inhibitor Rencofilstat Decreases HCV-induced Hepatocellular Carcinoma Independently of Its Antiviral Activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.19.553982. [PMID: 37645728 PMCID: PMC10462172 DOI: 10.1101/2023.08.19.553982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
There is an urgent need for the identification of new drugs that inhibit HCV-induced hepatocellular carcinoma (HCC). Our work demonstrates that cyclophilin inhibitors (CypI) represent such new drugs. We demonstrated that the non-immunosuppressive cyclosporine A (CsA) analog (CsAa) rencofilstat possesses dual therapeutic activities for the treatment of HCV infection and HCV-induced HCC. Specifically, we showed that HCV infection of humanized mice results in the progressive development of HCC. This was true for four genotypes tested (1 to 4). Remarkably, we demonstrated that rencofilstat inhibits the development of HCV-induced HCC in mice even when added 16 weeks post-infection when HCC is well established. Importantly, we showed that rencofilstat drastically reduces HCC progression independently of its anti-HCV activity. Indeed, the CypI rencofilstat inhibits HCC while other anti-HCV agents such as NS5A (NS5Ai) and NS5B (NS5Bi) fail to reduce HCC. In conclusion, this study shows for the first time that the CypI rencofilstat represents a potent therapeutic agent for the treatment of HCV-induced HCC.
Collapse
Affiliation(s)
- Winston Stauffer
- Department of Immunology & Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Michael Bobardt
- Department of Immunology & Microbiology, The Scripps Research Institute, La Jolla, California, USA
| | - Daren Ure
- Hepion Pharmaceuticals Inc., Edison, New Jersey, USA
| | - Robert Foster
- Hepion Pharmaceuticals Inc., Edison, New Jersey, USA
| | - Philippe Gallay
- Department of Immunology & Microbiology, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
7
|
Flaxman HA, Chrysovergi MA, Han H, Kabir F, Lister RT, Chang CF, Black KE, Lagares D, Woo CM. Sanglifehrin A mitigates multi-organ fibrosis in vivo by inducing secretion of the collagen chaperone cyclophilin B. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.09.531890. [PMID: 36945535 PMCID: PMC10028952 DOI: 10.1101/2023.03.09.531890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Pathological deposition and crosslinking of collagen type I by activated myofibroblasts drives progressive tissue fibrosis. Therapies that inhibit collagen synthesis by myofibroblasts have clinical potential as anti-fibrotic agents. Lysine hydroxylation by the prolyl-3-hydroxylase complex, comprised of cartilage associated protein, prolyl 3-hydroxylase 1, and cyclophilin B, is essential for collagen type I crosslinking and formation of stable fibers. Here, we identify the collagen chaperone cyclophilin B as a major cellular target of the macrocyclic natural product sanglifehrin A (SfA) using photo-affinity labeling and chemical proteomics. Our studies reveal a unique mechanism of action in which SfA binding to cyclophilin B in the endoplasmic reticulum (ER) induces the secretion of cyclophilin B to the extracellular space, preventing TGF-β1-activated myofibroblasts from synthesizing collagen type I in vitro without inhibiting collagen type I mRNA transcription or inducing ER stress. In addition, SfA prevents collagen type I secretion without affecting myofibroblast contractility or TGF-β1 signaling. In vivo, we provide chemical, molecular, functional, and translational evidence that SfA mitigates the development of lung and skin fibrosis in mouse models by inducing cyclophilin B secretion, thereby inhibiting collagen synthesis from fibrotic fibroblasts in vivo . Consistent with these findings in preclinical models, SfA reduces collagen type I secretion from fibrotic human lung fibroblasts and precision cut lung slices from patients with idiopathic pulmonary fibrosis, a fatal fibrotic lung disease with limited therapeutic options. Our results identify the primary liganded target of SfA in cells, the collagen chaperone cyclophilin B, as a new mechanistic target for the treatment of organ fibrosis.
Collapse
|
8
|
Targeting Artemisinin-Resistant Malaria by Repurposing the Anti-Hepatitis C Virus Drug Alisporivir. Antimicrob Agents Chemother 2022; 66:e0039222. [PMID: 36374050 PMCID: PMC9765015 DOI: 10.1128/aac.00392-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The emergence of Plasmodium falciparum resistance raises an urgent need to find new antimalarial drugs. Here, we report the rational repurposing of the anti-hepatitis C virus drug, alisporivir, a nonimmunosuppressive analog of cyclosporin A, against artemisinin-resistant strains of P. falciparum. In silico docking studies and molecular dynamic simulation predicted strong interaction of alisporivir with PfCyclophilin 19B, confirmed through biophysical assays with a Kd value of 354.3 nM. Alisporivir showed potent antimalarial activity against chloroquine-resistant (PfRKL-9 with resistance index [Ri] 2.14 ± 0.23) and artemisinin-resistant (PfKelch13R539T with Ri 1.15 ± 0.04) parasites. The Ri is defined as the ratio between the IC50 values of the resistant line to that of the sensitive line. To further investigate the mechanism involved, we analyzed the expression level of PfCyclophilin 19B in artemisinin-resistant P. falciparum (PfKelch13R539T). Semiquantitative real-time transcript, Western blot, and immunofluorescence analyses confirmed the overexpression of PfCyclophilin 19B in PfKelch13R539T. A 50% inhibitory concentration in the nanomolar range, together with the targeting of PfCyclophilin 19B, suggests that alisporivir can be used in combination with artemisinin. Since artemisinin resistance slows the clearance of ring-stage parasites, we performed a ring survival assay on artemisinin-resistant strain PfKelch13R539T and found significant decrease in parasite survival with alisporivir. Alisporivir was found to act synergistically with dihydroartemisinin and increase its efficacy. Furthermore, alisporivir exhibited antimalarial activity in vivo. Altogether, with the rational target-based Repurposing of alisporivir against malaria, our results support the hypothesis that targeting resistance mechanisms is a viable approach toward dealing with drug-resistant parasite.
Collapse
|
9
|
Entry inhibition of hepatitis B virus using cyclosporin O derivatives with peptoid side chain incorporation. Bioorg Med Chem 2022; 68:116862. [PMID: 35691131 DOI: 10.1016/j.bmc.2022.116862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 11/23/2022]
Abstract
Hepatitis B virus (HBV) infection is a serious worldwide health problem causing liver cirrhosis and hepatocellular carcinoma. The development of novel therapeutics targeting distinct steps of the HBV life cycle and combination therapy with approved drugs (i.e., nucleot(s)ides, interferon-α) are considered effective strategies for curing HBV. Among these strategies is the development of entry inhibitors that interfere with the host entry step of HBV to prevent viral infection and transmission. Herein, we generated a novel library of cyclosporin O (CsO) derivatives that incorporate peptoid side chains. Twenty-two CsO derivatives were evaluated for membrane permeability, cytotoxicity, and in vitro HBV entry inhibitory activity. The lead compound (i.e., compound 21) showed the greatest potency in the in vitro HBV entry inhibition assay (IC50 = 0.36 ± 0.01 μM) with minimal cytotoxicity. Our peptide-peptoid hybrid CsO scaffold can readily expand chemical diversity and is applicable for screening various targets requiring macrocyclic chemical entities.
Collapse
|
10
|
Banerjee D, Patra D, Sinha A, Roy S, Pant R, Sarmah R, Dutta R, Kanta Bhagabati S, Tikoo K, Pal D, Dasgupta S. Lipid-induced monokine cyclophilin-A promotes adipose tissue dysfunction implementing insulin resistance and type 2 diabetes in zebrafish and mice models of obesity. Cell Mol Life Sci 2022; 79:282. [PMID: 35511344 PMCID: PMC11072608 DOI: 10.1007/s00018-022-04306-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 04/01/2022] [Accepted: 04/11/2022] [Indexed: 11/28/2022]
Abstract
Several studies have implicated obesity-induced macrophage-adipocyte cross-talk in adipose tissue dysfunction and insulin resistance. However, the molecular cues involved in the cross-talk of macrophage and adipocyte causing insulin resistance are currently unknown. Here, we found that a lipid-induced monokine cyclophilin-A (CyPA) significantly attenuates adipocyte functions and insulin sensitivity. Targeted inhibition of CyPA in diet-induced obese zebrafish notably reduced adipose tissue inflammation and restored adipocyte function resulting in improvement of insulin sensitivity. Silencing of macrophage CyPA or pharmacological inhibition of CyPA by TMN355 effectively restored adipocytes' functions and insulin sensitivity. Interestingly, CyPA incubation markedly increased adipocyte inflammation along with an impairment of adipogenesis, however, mutation of its cognate receptor CD147 at P309A and G310A significantly waived CyPA's effect on adipocyte inflammation and its differentiation. Mechanistically, CyPA-CD147 interaction activates NF-κB signaling which promotes adipocyte inflammation by upregulating various pro-inflammatory cytokines gene expression and attenuates adipocyte differentiation by inhibiting PPARγ and C/EBPβ expression via LZTS2-mediated downregulation of β-catenin. Moreover, inhibition of CyPA or its receptor CD147 notably restored palmitate or CyPA-induced adipose tissue dysfunctions and insulin sensitivity. All these results indicate that obesity-induced macrophage-adipocyte cross-talk involving CyPA-CD147 could be a novel target for the management of insulin resistance and type 2 diabetes.
Collapse
Affiliation(s)
- Dipanjan Banerjee
- Metabolic Disease Biology Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, 784028, Assam, India
| | - Debarun Patra
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, 140001, Punjab, India
| | - Archana Sinha
- Metabolic Disease Biology Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, 784028, Assam, India
| | - Soumyajit Roy
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, 140001, Punjab, India
| | - Rajat Pant
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, 160062, India
| | - Raktim Sarmah
- Department of Aquatic Environment Management, College of Fisheries, Assam Agricultural University, Nagaon, 782103, Assam, India
| | - Rajdeep Dutta
- Department of Aquatic Environment Management, College of Fisheries, Assam Agricultural University, Nagaon, 782103, Assam, India
| | - Sarada Kanta Bhagabati
- Department of Aquatic Environment Management, College of Fisheries, Assam Agricultural University, Nagaon, 782103, Assam, India
| | - Kulbhushan Tikoo
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, 160062, India
| | - Durba Pal
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, 140001, Punjab, India.
| | - Suman Dasgupta
- Metabolic Disease Biology Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, 784028, Assam, India.
| |
Collapse
|
11
|
Mamatis JE, Pellizzari-Delano IE, Gallardo-Flores CE, Colpitts CC. Emerging Roles of Cyclophilin A in Regulating Viral Cloaking. Front Microbiol 2022; 13:828078. [PMID: 35242122 PMCID: PMC8886124 DOI: 10.3389/fmicb.2022.828078] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/26/2022] [Indexed: 12/24/2022] Open
Abstract
Cellular cyclophilins (Cyps) such as cyclophilin A (CypA) have emerged as key players at the virus-host interface. As host factors required for the replication of many unrelated viruses, including human immunodeficiency virus (HIV), hepatitis C virus (HCV) and coronaviruses (CoVs), Cyps are attractive targets for antiviral therapy. However, a clear understanding of how these viruses exploit Cyps to promote their replication has yet to be elucidated. Recent findings suggest that CypA contributes to cloaking of viral replication intermediates, an evasion strategy that prevents detection of viral nucleic acid by innate immune sensors. Furthermore, Cyps are emerging to have roles in regulation of cellular antiviral signaling pathways. Recruitment of Cyps by viral proteins may interfere with their ability to regulate these signaling factors. Consistent with disruption of viral cloaking and innate immune evasion, treatment with Cyp inhibitors such as cyclosporine A (CsA) restores antiviral innate immunity and induces expression of a subset of antiviral genes that restrict viral infection, which may help to explain the broad antiviral spectrum of CsA. In this review, we provide an overview of the roles of CypA in viral cloaking and evasion of innate immunity, focusing on the underlying mechanisms and new perspectives for antiviral therapies.
Collapse
Affiliation(s)
- John E Mamatis
- Department of Biomedical and Molecular Sciences, Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | - Isabella E Pellizzari-Delano
- Department of Biomedical and Molecular Sciences, Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | - Carla E Gallardo-Flores
- Department of Biomedical and Molecular Sciences, Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| | - Che C Colpitts
- Department of Biomedical and Molecular Sciences, Faculty of Health Sciences, Queen's University, Kingston, ON, Canada
| |
Collapse
|
12
|
Corbett KM, Ford L, Warren DB, Pouton CW, Chalmers DK. Cyclosporin Structure and Permeability: From A to Z and Beyond. J Med Chem 2021; 64:13131-13151. [PMID: 34478303 DOI: 10.1021/acs.jmedchem.1c00580] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cyclosporins are natural or synthetic undecapeptides with a wide range of actual and potential pharmaceutical applications. Several members of the cyclosporin compound family have remarkably high passive membrane permeabilities that are not well-described by simple structural metrics. Here we review experimental studies of cyclosporin structure and permeability, including cyclosporin-metal complexes. We also discuss models for the conformation-dependent permeability of cyclosporins and similar compounds. Finally, we identify current knowledge gaps in the literature and provide recommendations regarding future avenues of exploration.
Collapse
Affiliation(s)
- Karen M Corbett
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Leigh Ford
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Dallas B Warren
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Colin W Pouton
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - David K Chalmers
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| |
Collapse
|
13
|
Rigotto G, Zentilin L, Pozzan T, Basso E. Effects of Mild Excitotoxic Stimulus on Mitochondria Ca 2+ Handling in Hippocampal Cultures of a Mouse Model of Alzheimer's Disease. Cells 2021; 10:cells10082046. [PMID: 34440815 PMCID: PMC8394681 DOI: 10.3390/cells10082046] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/02/2021] [Accepted: 08/06/2021] [Indexed: 01/19/2023] Open
Abstract
In Alzheimer’s disease (AD), the molecular mechanisms involved in the neurodegeneration are still incompletely defined, though this aspect is crucial for a better understanding of the malady and for devising effective therapies. Mitochondrial dysfunctions and altered Ca2+ signaling have long been implicated in AD, though it is debated whether these events occur early in the course of the pathology, or whether they develop at late stages of the disease and represent consequences of different alterations. Mitochondria are central to many aspects of cellular metabolism providing energy, lipids, reactive oxygen species, signaling molecules for cellular quality control, and actively shaping intracellular Ca2+ signaling, modulating the intensity and duration of the signal itself. Abnormalities in the ability of mitochondria to take up and subsequently release Ca2+ could lead to changes in the metabolism of the organelle, and of the cell as a whole, that eventually result in cell death. We sought to investigate the role of mitochondria and Ca2+ signaling in a model of Familial Alzheimer’s disease and found early alterations in mitochondria physiology under stressful condition, namely, reduced maximal respiration, decreased ability to sustain membrane potential, and a slower return to basal matrix Ca2+ levels after a mild excitotoxic stimulus. Treatment with an inhibitor of the permeability transition pore attenuated some of these mitochondrial disfunctions and may represent a promising tool to ameliorate mitochondria and cellular functioning in AD and prevent or slow down cell loss in the disease.
Collapse
Affiliation(s)
- Giulia Rigotto
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (G.R.); (T.P.)
| | - Lorena Zentilin
- International Centre for Genetic Engineering and Biotechnology (ICGEB), 34149 Trieste, Italy;
| | - Tullio Pozzan
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (G.R.); (T.P.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
- Venetian Institute of Molecular Medicine (VIMM), 35131 Padua, Italy
| | - Emy Basso
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy; (G.R.); (T.P.)
- Neuroscience Institute, National Research Council (CNR), 35131 Padua, Italy
- Correspondence:
| |
Collapse
|
14
|
Chang C, Flaxman HA, Woo CM. Enantioselective Synthesis and Biological Evaluation of Sanglifehrin A and B and Analogs. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202103022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Chia‐Fu Chang
- Department of Chemistry and Chemical Biology Harvard University 12 Oxford St Cambridge MA 02138 USA
| | - Hope A. Flaxman
- Department of Chemistry and Chemical Biology Harvard University 12 Oxford St Cambridge MA 02138 USA
| | - Christina M. Woo
- Department of Chemistry and Chemical Biology Harvard University 12 Oxford St Cambridge MA 02138 USA
| |
Collapse
|
15
|
Lee D, Lee S, Choi J, Song YK, Kim MJ, Shin DS, Bae MA, Kim YC, Park CJ, Lee KR, Choi JH, Seo J. Interplay among Conformation, Intramolecular Hydrogen Bonds, and Chameleonicity in the Membrane Permeability and Cyclophilin A Binding of Macrocyclic Peptide Cyclosporin O Derivatives. J Med Chem 2021; 64:8272-8286. [PMID: 34096287 DOI: 10.1021/acs.jmedchem.1c00211] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A macrocyclic peptide scaffold with well-established structure-property relationship is desirable for tackling undruggable targets. Here, we adopted a natural macrocycle, cyclosporin O (CsO) and its derivatives (CP1-3), and evaluated the impact of conformation on membrane permeability, cyclophilin A (CypA) binding, and the pharmacokinetic (PK) profile. In nonpolar media, CsO showed a similar conformation to cyclosporin A (CsA), a well-known chameleonic macrocycle, but less chameleonic behavior in a polar environment. The weak chameleonicity of CsO resulted in decreased membrane permeability; however, the more rigid conformation of CsO was not detrimental to its PK profile. CsO exhibited a higher plasma concentration than CsA, which resulted from minimal CypA binding and lower accumulation in red blood cells and moderate oral bioavailability (F = 12%). Our study aids understanding of CsO, a macrocyclic peptide that is less explored than CsA but with greater potential for diversity generation and rational design.
Collapse
Affiliation(s)
- Dongjae Lee
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Sungjin Lee
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Jieun Choi
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Yoo-Kyung Song
- Laboratory of Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Republic of Korea
| | - Min Ju Kim
- Laboratory of Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Republic of Korea
| | - Dae-Seop Shin
- Bio Platform Technology Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Myung Ae Bae
- Bio Platform Technology Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Republic of Korea
| | - Yong-Chul Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Chin-Ju Park
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Kyeong-Ryoon Lee
- Laboratory of Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 28116, Republic of Korea
| | - Jun-Ho Choi
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Jiwon Seo
- Department of Chemistry, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| |
Collapse
|
16
|
Chang CF, Flaxman HA, Woo CM. Enantioselective Synthesis and Biological Evaluation of Sanglifehrin A and B and Analogs. Angew Chem Int Ed Engl 2021; 60:17045-17052. [PMID: 34014025 DOI: 10.1002/anie.202103022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/03/2021] [Indexed: 01/02/2023]
Abstract
Sanglifehrin A and B are immunosuppressive macrocyclic natural products endowed with and differentiated by a unique spirocyclic lactam. Herein, we report an enantioselective total synthesis and biological evaluation of sanglifehrin A and B and analogs. Access to the spirocyclic lactam was achieved through convergent assembly of a key pyranone intermediate followed by a stereo-controlled spirocyclization. The 22-membered macrocyclic core was synthesized by ring-closing metathesis in the presence of 2,6-bis(trifluoromethyl) benzeneboronic acid (BFBB). The spirocyclic lactam and macrocycle fragments were united by a Stille coupling to furnish sanglifehrin A and B. Additional sanglifehrin B analogs with variation at the C40 position were additionally prepared. Biological evaluation revealed that the 2-CF3 analog of sanglifehrin B exhibited higher anti-proliferative activity than the natural products sanglifehrin A and B in Jurkat cells. Both natural products induced higher-order homodimerization of cyclophilin A (CypA), but only sanglifehrin A promoted CypA complexation with inosine-5'-monophosphate dehydrogenase 2 (IMPDH2). The synthesis reported herein will enable further evaluation of the spirolactam and its contribution to sanglifehrin-dependent immunosuppressive activity.
Collapse
Affiliation(s)
- Chia-Fu Chang
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St, Cambridge, MA, 02138, USA
| | - Hope A Flaxman
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St, Cambridge, MA, 02138, USA
| | - Christina M Woo
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford St, Cambridge, MA, 02138, USA
| |
Collapse
|
17
|
Abstract
The non-immune-suppressive cyclophilin inhibitor CRV431 is a clinical candidate to cure nonalcoholic steatohepatitis (NASH) and has the potential to treat liver fibrosis and cancer incidence. Herein we report a concise chemical semisynthesis of CRV431 in four steps from the commercially available cyclosporine, featuring in this the flow-chemistry-based methylenation an intermolecular ring-closing metathesis and a Rh-catalyzed diastereoselective hydrogenation.
Collapse
Affiliation(s)
- Feng-Xia Li
- State Key Laboratory of Chemical Oncogenomics and Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Qing-Zhou Zhang
- State Key Laboratory of Chemical Oncogenomics and Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Shi-Jun Li
- Green Catalysis Center and College of Chemistry, Zhengzhou University, 100 Science Avenue, Zhengzhou, Henan 450001, P. R. China
| | - Guang Lin
- State Key Laboratory of Chemical Oncogenomics and Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Xiang-Yu Huo
- State Key Laboratory of Chemical Oncogenomics and Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Yu Lan
- Green Catalysis Center and College of Chemistry, Zhengzhou University, 100 Science Avenue, Zhengzhou, Henan 450001, P. R. China.,School of Chemistry and Chemical Engineering, and Chongqing Key Laboratory of Theoretical and Computational Chemistry, Chongqing University, Chongqing 400030, P. R. China
| | - Zhen Yang
- State Key Laboratory of Chemical Oncogenomics and Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China.,Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education and Beijing National Laboratory for Molecular Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China.,Shenzhen Bay Laboratory, Shenzhen 518055, China
| |
Collapse
|
18
|
Hakim S, Craig JM, Koblinski JE, Clevenger CV. Inhibition of the Activity of Cyclophilin A Impedes Prolactin Receptor-Mediated Signaling, Mammary Tumorigenesis, and Metastases. iScience 2020; 23:101581. [PMID: 33083747 PMCID: PMC7549119 DOI: 10.1016/j.isci.2020.101581] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/27/2020] [Accepted: 09/15/2020] [Indexed: 01/12/2023] Open
Abstract
Prolactin (PRL) and its receptor (PRLr) play important roles in the pathogenesis of breast cancer. Cyclophilin A (CypA) is a cis-trans peptidyl-prolyl isomerase (PPI) that is constitutively associated with the PRLr and facilitates the activation of the tyrosine kinase Jak2. Treatment with the non-immunosuppressive prolyl isomerase inhibitor NIM811 or CypA short hairpin RNA inhibited PRL-stimulated signaling, breast cancer cell growth, and migration. Transcriptomic analysis revealed that NIM811 inhibited two-thirds of the top 50 PRL-induced genes and a reduction in gene pathways associated with cancer cell signaling. In vivo treatment of NIM811 in a TNBC xenograft lessened primary tumor growth and induced central tumor necrosis. Deletion of CypA in the MMTV-PyMT mouse model demonstrated inhibition of tumorigenesis with significant reduction in lung and lymph node metastasis. The regulation of PRLr/Jak2-mediated biology by NIM811 demonstrates that a non-immunosuppressive prolyl isomerase inhibitor can function as a potential breast cancer therapeutic. CypA inhibition or knockdown blocks breast cancer cell signaling, growth, and migration NIM811 inhibited PRL-induced genes and gene pathways relevant to cancer signaling Deletion of CypA has shown reduction in tumorigenesis and metastasis in mice
Collapse
Affiliation(s)
- Shawn Hakim
- Department of Pathology, Virginia Commonwealth University, 1101 E. Marshall St, Sanger 4-006A, Richmond, VA 23298, USA.,Massey Cancer Center, Richmond, VA 23298, USA.,Wright Center for Clinical and Translational Sciences, Richmond, VA 23298, USA
| | - Justin M Craig
- Department of Pathology, Virginia Commonwealth University, 1101 E. Marshall St, Sanger 4-006A, Richmond, VA 23298, USA.,Massey Cancer Center, Richmond, VA 23298, USA.,Wright Center for Clinical and Translational Sciences, Richmond, VA 23298, USA
| | - Jennifer E Koblinski
- Department of Pathology, Virginia Commonwealth University, 1101 E. Marshall St, Sanger 4-006A, Richmond, VA 23298, USA.,Massey Cancer Center, Richmond, VA 23298, USA
| | - Charles V Clevenger
- Department of Pathology, Virginia Commonwealth University, 1101 E. Marshall St, Sanger 4-006A, Richmond, VA 23298, USA.,Massey Cancer Center, Richmond, VA 23298, USA
| |
Collapse
|
19
|
Liu C, von Brunn A, Zhu D. Cyclophilin A and CD147: novel therapeutic targets for the treatment of COVID-19. MEDICINE IN DRUG DISCOVERY 2020; 7:100056. [PMID: 32835213 PMCID: PMC7364167 DOI: 10.1016/j.medidd.2020.100056] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/05/2020] [Accepted: 07/05/2020] [Indexed: 02/07/2023] Open
Abstract
The outbreak of pneumonia caused by a new coronavirus (SARS-CoV-2) occurred in December 2019, and spread rapidly throughout the world. There have been other severe coronavirus outbreaks worldwide, namely, severe acute respiratory syndrome (SARS-CoV) and Middle East respiratory syndrome (MERS-CoV). Because the genetic diversity of coronaviruses renders the design of vaccines complicated, broad spectrum-anti-coronavirus drugs have become a critical approach to control the coronavirus epidemic. Cyclophilin A is an important protein needed for coronavirus replication, and its inhibitor cyclosporine A has the ability to suppress coronavirus on a broad spectrum. CD147-S protein was found to be one route by which SARS-CoV-2 invades host cells, while CD147 was found to play a functional role in facilitating the infection of host cells by SARS-CoV. The CyPA/CD147 interaction may play a critical role in the ability of the SARS-CoV-2 virus to enter the host cells. However, cyclosporine A has immunosuppressive effects, so the conditions for its use as an antiviral drug are limited. As a result, cyclosporine A analogues without immunosuppressive side effects have attracted lots of interest. This review primarily discusses the drug development prospects of cyclophilin A as a therapeutic target for the treatment of coronavirus infection, especially coronavirus disease 2019 (COVID-19), and non-immunosuppressive cyclosporine analogues.
Collapse
Affiliation(s)
- Chenglong Liu
- School of Pharmacy, Fudan University, Shanghai, China, 201203
| | - Albrecht von Brunn
- Max-von-Pettenkofer Institute, Ludwig-Maximilians-University Munich and German Center for Infection Research, (DZIF), partner site Munich, 80336 Munich, Germany
| | - Di Zhu
- School of Pharmacy, Fudan University, Shanghai, China, 201203,Fudan affiliated Pudong Hospital, Fudan University, Shanghai, China 201100,Corresponding author at: School of Pharmacy, Fudan University, Shanghai, China 201203.
| |
Collapse
|
20
|
Scheuplein NJ, Bzdyl NM, Kibble EA, Lohr T, Holzgrabe U, Sarkar-Tyson M. Targeting Protein Folding: A Novel Approach for the Treatment of Pathogenic Bacteria. J Med Chem 2020; 63:13355-13388. [PMID: 32786507 DOI: 10.1021/acs.jmedchem.0c00911] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Infectious diseases are a major cause of morbidity and mortality worldwide, exacerbated by increasing antibiotic resistance in many bacterial species. The development of drugs with new modes of action is essential. A leading strategy is antivirulence, with the aim to target bacterial proteins that are important in disease causation and progression but do not affect growth, resulting in reduced selective pressure for resistance. Immunophilins, a superfamily of peptidyl-prolyl cis-trans isomerase (PPIase) enzymes have been shown to be important for virulence in a broad-spectrum of pathogenic bacteria. This Perspective will provide an overview of the recent advances made in understanding the role of each immunophilin family, cyclophilins, FK506 binding proteins (FKBPs), and parvulins in bacteria. Inhibitor design and medicinal chemistry strategies for development of novel drugs against bacterial FKBPs will be discussed. Furthermore, drugs against human cyclophilins and parvulins will be reviewed in their current indication as antiviral and anticancer therapies.
Collapse
Affiliation(s)
- Nicolas J Scheuplein
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Nicole M Bzdyl
- Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, 6009 Perth, Australia
| | - Emily A Kibble
- Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, 6009 Perth, Australia.,School of Veterinary and Life Sciences, Murdoch University, 6150 Murdoch, Australia
| | - Theresa Lohr
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Ulrike Holzgrabe
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Mitali Sarkar-Tyson
- Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, University of Western Australia, 6009 Perth, Australia
| |
Collapse
|
21
|
Brayden D, Hill T, Fairlie D, Maher S, Mrsny R. Systemic delivery of peptides by the oral route: Formulation and medicinal chemistry approaches. Adv Drug Deliv Rev 2020; 157:2-36. [PMID: 32479930 DOI: 10.1016/j.addr.2020.05.007] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023]
Abstract
In its 33 years, ADDR has published regularly on the po5tential of oral delivery of biologics especially peptides and proteins. In the intervening period, analysis of the preclinical and clinical trial failures of many purported platform technologies has led to reflection on the true status of the field and reigning in of expectations. Oral formulations of semaglutide, octreotide, and salmon calcitonin have completed Phase III trials, with oral semaglutide being approved by the FDA in 2019. The progress made with oral peptide formulations based on traditional permeation enhancers is against a background of low and variable oral bioavailability values of ~1%, leading to a current perception that only potent peptides with a viable cost of synthesis can be realistically considered. Desirable features of candidates should include a large therapeutic index, some stability in the GI tract, a long elimination half-life, and a relatively low clearance rate. Administration in nanoparticle formats have largely disappointed, with few prototypes reaching clinical trials: insufficient particle loading, lack of controlled release, low epithelial particle uptake, and lack of scalable synthesis being the main reasons for discontinuation. Disruptive technologies based on engineered devices promise improvements, but scale-up and toxicology aspects are issues to address. In parallel, medicinal chemists are synthesizing stable hydrophobic macrocyclic candidate peptides of lower molecular weight and with potential for greater oral bioavailability than linear peptides, but perhaps without the same requirement for elaborate drug delivery systems. In summary, while there have been advances in understanding the limitations of peptides for oral delivery, low membrane permeability, metabolism, and high clearance rates continue to hamper progress.
Collapse
|
22
|
Winquist RJ, Gribkoff VK. Targeting putative components of the mitochondrial permeability transition pore for novel therapeutics. Biochem Pharmacol 2020; 177:113995. [PMID: 32339494 DOI: 10.1016/j.bcp.2020.113995] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 04/22/2020] [Indexed: 12/14/2022]
Abstract
Few discoveries have influenced drug discovery programs more than the finding that mitochondrial membranes undergo swings in permeability in response to cellular perturbations. The conductor of these permeability changes is the aptly named mitochondrial permeability transition pore which, although not yet precisely defined, is comprised of several integral proteins that differentially act to regulate the flux of ions, proteins and metabolic byproducts during the course of cellular physiological functions but also pathophysiological insults. Pursuit of the pore's exact identity remains a topic of keen interest, but decades of research have unearthed provocative functions for the integral proteins leading to their evaluation to develop novel therapeutics for a wide range of clinical indications. Chief amongst these targeted, integral proteins have been the Voltage Dependent Anion Channel (VDAC) and the F1FO ATP synthase. Research associated with the roles and ligands of VDAC has been extensive and we will expand upon 3 examples of ligand:VDAC interactions for consideration of drug discovery projects: Tubulin:VDAC1, Hexokinase I/II:VDAC1 and olesoxime:VDAC1. The discoveries that cyclosporine blocks mitochondrial permeability transition via binding to cyclophilin D, and that cyclophilin D is an important component of F1FO ATP synthase, has heightened interest in the F1FO ATP synthase as a focal point for drug discovery, and we will discuss 2 plausible campaigns associated with disease indications. To date no drug has emerged from prospective targeting these integral proteins; however, continued exploration such as the approaches suggested in this Commentary will increase the likelihood of providing important therapeutics for severely unmet medical needs.
Collapse
Affiliation(s)
- Raymond J Winquist
- Alkermes Pharmaceuticals Inc, 852 Winter Street, Waltham MA 02451, United States.
| | - Valentin K Gribkoff
- Yale University School of Medicine, Department of Internal Medicine, 333 Cedar St., New Haven, CT 06510, United States; TheraStat LLC, 44 Kings Grant Rd., Weston, MA 02493, United States
| |
Collapse
|
23
|
Juárez-Jiménez J, Gupta AA, Karunanithy G, Mey ASJS, Georgiou C, Ioannidis H, De Simone A, Barlow PN, Hulme AN, Walkinshaw MD, Baldwin AJ, Michel J. Dynamic design: manipulation of millisecond timescale motions on the energy landscape of cyclophilin A. Chem Sci 2020; 11:2670-2680. [PMID: 34084326 PMCID: PMC8157532 DOI: 10.1039/c9sc04696h] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 01/14/2020] [Indexed: 12/21/2022] Open
Abstract
Proteins need to interconvert between many conformations in order to function, many of which are formed transiently, and sparsely populated. Particularly when the lifetimes of these states approach the millisecond timescale, identifying the relevant structures and the mechanism by which they interconvert remains a tremendous challenge. Here we introduce a novel combination of accelerated MD (aMD) simulations and Markov state modelling (MSM) to explore these 'excited' conformational states. Applying this to the highly dynamic protein CypA, a protein involved in immune response and associated with HIV infection, we identify five principally populated conformational states and the atomistic mechanism by which they interconvert. A rational design strategy predicted that the mutant D66A should stabilise the minor conformations and substantially alter the dynamics, whereas the similar mutant H70A should leave the landscape broadly unchanged. These predictions are confirmed using CPMG and R1ρ solution state NMR measurements. By efficiently exploring functionally relevant, but sparsely populated conformations with millisecond lifetimes in silico, our aMD/MSM method has tremendous promise for the design of dynamic protein free energy landscapes for both protein engineering and drug discovery.
Collapse
Affiliation(s)
- Jordi Juárez-Jiménez
- EaStCHEM School of Chemistry, University of Edinburgh David Brewster Road Edinburgh EH9 3FJ UK
| | - Arun A Gupta
- EaStCHEM School of Chemistry, University of Edinburgh David Brewster Road Edinburgh EH9 3FJ UK
| | - Gogulan Karunanithy
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford South Parks Road Oxford OX1 3QZ UK
| | - Antonia S J S Mey
- EaStCHEM School of Chemistry, University of Edinburgh David Brewster Road Edinburgh EH9 3FJ UK
| | - Charis Georgiou
- EaStCHEM School of Chemistry, University of Edinburgh David Brewster Road Edinburgh EH9 3FJ UK
| | - Harris Ioannidis
- EaStCHEM School of Chemistry, University of Edinburgh David Brewster Road Edinburgh EH9 3FJ UK
| | - Alessio De Simone
- EaStCHEM School of Chemistry, University of Edinburgh David Brewster Road Edinburgh EH9 3FJ UK
| | - Paul N Barlow
- EaStCHEM School of Chemistry, University of Edinburgh David Brewster Road Edinburgh EH9 3FJ UK
| | - Alison N Hulme
- EaStCHEM School of Chemistry, University of Edinburgh David Brewster Road Edinburgh EH9 3FJ UK
| | - Malcolm D Walkinshaw
- School of Biological Sciences Michael Swann Building, Max Born Crescent Edinburgh EH9 3BF UK
| | - Andrew J Baldwin
- Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford South Parks Road Oxford OX1 3QZ UK
| | - Julien Michel
- EaStCHEM School of Chemistry, University of Edinburgh David Brewster Road Edinburgh EH9 3FJ UK
| |
Collapse
|
24
|
Ure DR, Trepanier DJ, Mayo PR, Foster RT. Cyclophilin inhibition as a potential treatment for nonalcoholic steatohepatitis (NASH). Expert Opin Investig Drugs 2019; 29:163-178. [PMID: 31868526 DOI: 10.1080/13543784.2020.1703948] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Daren R. Ure
- Hepion Pharmaceuticals Inc, Edmonton, AB, Canada
| | | | | | | |
Collapse
|
25
|
Kuo J, Bobardt M, Chatterji U, Mayo PR, Trepanier DJ, Foster RT, Gallay P, Ure DR. A Pan-Cyclophilin Inhibitor, CRV431, Decreases Fibrosis and Tumor Development in Chronic Liver Disease Models. J Pharmacol Exp Ther 2019; 371:231-241. [PMID: 31406003 PMCID: PMC6815936 DOI: 10.1124/jpet.119.261099] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022] Open
Abstract
Previous studies show that cyclophilins contribute to many pathologic processes, and cyclophilin inhibitors demonstrate therapeutic activities in many experimental models. However, no drug with cyclophilin inhibition as the primary mode of action has advanced completely through clinical development to market. In this study, we present findings on the cyclophilin inhibitor, CRV431, that highlight its potential as a drug candidate for chronic liver diseases. CRV431 was found to potently inhibit all cyclophilin isoforms tested—A, B, D, and G. Inhibitory constant or IC50 values ranged from 1 to 7 nM, which was up to 13 times more potent than the parent compound, cyclosporine A (CsA), from which CRV431 was derived. Other CRV431 advantages over CsA as a nontransplant drug candidate were significantly diminished immunosuppressive activity, less drug transporter inhibition, and reduced cytotoxicity potential. Oral dosing to mice and rats led to good blood exposures and a 5- to 15-fold accumulation of CRV431 in liver compared with blood concentrations across a wide range of CRV431 dosing levels. Most importantly, CRV431 decreased liver fibrosis in a 6-week carbon tetrachloride model and in a mouse model of nonalcoholic steatohepatitis (NASH). Additionally, CRV431 administration during a late, oncogenic stage of the NASH disease model resulted in a 50% reduction in the number and size of liver tumors. These findings are consistent with CRV431 targeting fibrosis and cancer through multiple, cyclophilin-mediated mechanisms and support the development of CRV431 as a safe and effective drug candidate for liver diseases.
Collapse
Affiliation(s)
- Joseph Kuo
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, California (J.K., M.B., U.C., P.G.); and Hepion Pharmaceuticals, Edison, New Jersey (P.R.M., D.J.T., R.T.F., D.R.U.)
| | - Michael Bobardt
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, California (J.K., M.B., U.C., P.G.); and Hepion Pharmaceuticals, Edison, New Jersey (P.R.M., D.J.T., R.T.F., D.R.U.)
| | - Udayan Chatterji
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, California (J.K., M.B., U.C., P.G.); and Hepion Pharmaceuticals, Edison, New Jersey (P.R.M., D.J.T., R.T.F., D.R.U.)
| | - Patrick R Mayo
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, California (J.K., M.B., U.C., P.G.); and Hepion Pharmaceuticals, Edison, New Jersey (P.R.M., D.J.T., R.T.F., D.R.U.)
| | - Daniel J Trepanier
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, California (J.K., M.B., U.C., P.G.); and Hepion Pharmaceuticals, Edison, New Jersey (P.R.M., D.J.T., R.T.F., D.R.U.)
| | - Robert T Foster
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, California (J.K., M.B., U.C., P.G.); and Hepion Pharmaceuticals, Edison, New Jersey (P.R.M., D.J.T., R.T.F., D.R.U.)
| | - Philippe Gallay
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, California (J.K., M.B., U.C., P.G.); and Hepion Pharmaceuticals, Edison, New Jersey (P.R.M., D.J.T., R.T.F., D.R.U.)
| | - Daren R Ure
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, California (J.K., M.B., U.C., P.G.); and Hepion Pharmaceuticals, Edison, New Jersey (P.R.M., D.J.T., R.T.F., D.R.U.)
| |
Collapse
|
26
|
Peruzzi MT, Gallou F, Lee SJ, Gagné MR. Site Selective Amide Reduction of Cyclosporine A Enables Diverse Derivation of an Important Cyclic Peptide. Org Lett 2019; 21:3451-3455. [PMID: 30993986 DOI: 10.1021/acs.orglett.9b01245] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Site selective amide reductions of the cyclic undecapeptide, cyclosporine A, have been developed using the combination of a heteroleptic borane catalyst and a silane reductant. Tertiary silane Me2EtSiH provides two unique cyclosporine A derivatives, one of which can be readily diversified in subsequent reactions. The secondary silane Et2SiH2 enables divergent reactivity that uses a free hydroxyl group to direct the reduction. The transient O-silyl hemiaminal intermediate of this reduction can additionally be trapped by reducing to the amine or by reductive cyanation.
Collapse
Affiliation(s)
- Michael T Peruzzi
- Caudill Laboratories, Department of Chemistry , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599-3290 , United States
| | - Fabrice Gallou
- Chemical and Analytical Development - Novartis Pharma AG , Basel , Switzerland
| | - Stephen J Lee
- U.S. Army Research Office , P.O. Box 12211, Research Triangle Park , North Carolina 27709 , United States
| | - Michel R Gagné
- Caudill Laboratories, Department of Chemistry , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599-3290 , United States
| |
Collapse
|
27
|
De Simone A, Georgiou C, Ioannidis H, Gupta AA, Juárez-Jiménez J, Doughty-Shenton D, Blackburn EA, Wear MA, Richards JP, Barlow PN, Carragher N, Walkinshaw MD, Hulme AN, Michel J. A computationally designed binding mode flip leads to a novel class of potent tri-vector cyclophilin inhibitors. Chem Sci 2019; 10:542-547. [PMID: 30746096 PMCID: PMC6335623 DOI: 10.1039/c8sc03831g] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/14/2018] [Indexed: 12/27/2022] Open
Abstract
Cyclophilins (Cyps) are a major family of drug targets that are challenging to prosecute with small molecules because the shallow nature and high degree of conservation of the active site across human isoforms offers limited opportunities for potent and selective inhibition. Herein a computational approach based on molecular dynamics simulations and free energy calculations was combined with biophysical assays and X-ray crystallography to explore a flip in the binding mode of a reported urea-based Cyp inhibitor. This approach enabled access to a distal pocket that is poorly conserved among key Cyp isoforms, and led to the discovery of a new family of sub-micromolar cell-active inhibitors that offer unprecedented opportunities for the development of next-generation drug therapies based on Cyp inhibition. The computational approach is applicable to a broad range of organic functional groups and could prove widely enabling in molecular design.
Collapse
Affiliation(s)
- Alessio De Simone
- University of Edinburgh , Joseph Black Building, King's Buildings, David Brewster Road , Edinburgh , Scotland EH9 3FJ , UK .
| | - Charis Georgiou
- University of Edinburgh , Joseph Black Building, King's Buildings, David Brewster Road , Edinburgh , Scotland EH9 3FJ , UK .
| | - Harris Ioannidis
- University of Edinburgh , Joseph Black Building, King's Buildings, David Brewster Road , Edinburgh , Scotland EH9 3FJ , UK .
| | - Arun A Gupta
- University of Edinburgh , Joseph Black Building, King's Buildings, David Brewster Road , Edinburgh , Scotland EH9 3FJ , UK .
| | - Jordi Juárez-Jiménez
- University of Edinburgh , Joseph Black Building, King's Buildings, David Brewster Road , Edinburgh , Scotland EH9 3FJ , UK .
| | - Dahlia Doughty-Shenton
- Edinburgh Phenotypic Assay Centre , University of Edinburgh , Queen's Medical Research Institute , Little France Cres , Edinburgh , Scotland EH16 4TJ , UK
| | - Elizabeth A Blackburn
- The Edinburgh Protein Production Facility (EPPF) , University of Edinburgh , Level 3 Michael Swann Building, King's Buildings, Max Born Crescent , Edinburgh , Scotland EH9 3BF , UK
| | - Martin A Wear
- The Edinburgh Protein Production Facility (EPPF) , University of Edinburgh , Level 3 Michael Swann Building, King's Buildings, Max Born Crescent , Edinburgh , Scotland EH9 3BF , UK
| | - Jonathan P Richards
- University of Edinburgh , Joseph Black Building, King's Buildings, David Brewster Road , Edinburgh , Scotland EH9 3FJ , UK .
| | - Paul N Barlow
- University of Edinburgh , Joseph Black Building, King's Buildings, David Brewster Road , Edinburgh , Scotland EH9 3FJ , UK .
| | - Neil Carragher
- Cancer Research UK Edinburgh Centre , University of Edinburgh , MRC Institute of Genetics and Molecular Medicine , Crewe Road South , Edinburgh , Scotland EH4 2XR , UK
| | - Malcolm D Walkinshaw
- University of Edinburgh , Michael Swann Building, Max Born Crescent , Edinburgh , Scotland EH9 3BF , UK
| | - Alison N Hulme
- University of Edinburgh , Joseph Black Building, King's Buildings, David Brewster Road , Edinburgh , Scotland EH9 3FJ , UK .
| | - Julien Michel
- University of Edinburgh , Joseph Black Building, King's Buildings, David Brewster Road , Edinburgh , Scotland EH9 3FJ , UK .
| |
Collapse
|
28
|
Blanco MJ. Building upon Nature's Framework: Overview of Key Strategies Toward Increasing Drug-Like Properties of Natural Product Cyclopeptides and Macrocycles. Methods Mol Biol 2019; 2001:203-233. [PMID: 31134573 DOI: 10.1007/978-1-4939-9504-2_10] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The pharmaceutical industry has focused mainly in the development of small-molecule entities intended for oral administration for the past decades. As a result, the majority of existing drugs address only a narrow range of biological targets. In the era of post-genomics, transcriptomics, and proteomics, there is an increasing interest on larger modulators of proteins that can span larger surfaces, access new therapeutic mechanisms of action, and provide greater target specificity. Traditional drug-like molecules developed using "rule-of-five" (Ro5) guidelines have been proven ineffective against a variety of challenging targets, such as protein-protein interactions, nucleic acid complexes, and antibacterial modalities. However, natural products are known to be effective at modulating such targets, leading to a renewed focus by medicinal chemists on investigating underrepresented chemical scaffolds associated with natural products. Here we describe recent efforts toward identification of novel natural cyclopeptides and macrocycles as well as selected medicinal chemistry strategies to increase drug-like properties or further exploration of their activity.
Collapse
|
29
|
Dujardin M, Bouckaert J, Rucktooa P, Hanoulle X. X-ray structure of alisporivir in complex with cyclophilin A at 1.5 Å resolution. Acta Crystallogr F Struct Biol Commun 2018; 74:583-592. [PMID: 30198892 PMCID: PMC6130424 DOI: 10.1107/s2053230x18010415] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 07/18/2018] [Indexed: 12/14/2022] Open
Abstract
Alisporivir (ALV) is an 11-amino-acid hydrophobic cyclic peptide with N-methyl-D-alanine and N-ethyl-L-valine (NEV) residues at positions 3 and 4, respectively. ALV is a non-immunosuppressive cyclosporin A (CsA) derivative. This inhibitor targets cyclophilins (Cyps), a family of proteins with peptidyl-prolyl cis/trans isomerase enzymatic activity. Cyps act as protein chaperones and are involved in numerous cellular functions. Moreover, Cyps have been shown to be an essential cofactor for the replication of many viruses, including Hepatitis C virus and Human immunodeficiency virus, and have also been shown to be involved in mitochondrial diseases. For these reasons, cyclophilins represent an attractive drug target. The structure of ALV in complex with cyclophilin A (CypA), the most abundant Cyp in humans, has been determined at 1.5 Å resolution. This first structure of the CypA-ALV complex shows that the binding of ALV is highly similar to that of CsA. The high resolution allowed the unambiguous determination of the conformations of residues 3 and 4 in ALV when bound to its target. In particular, the side-chain conformation of NEV4 precludes the interaction of the CypA-ALV complex with calcineurin, a cellular protein phosphatase involved in the immune response, which explains the non-immunosuppressive property of ALV. This study provides detailed molecular insights into the CypA-ALV interaction.
Collapse
Affiliation(s)
- Marie Dujardin
- UGSF–UMR8576, University of Lille, CNRS, F-59000 Lille, France
| | - Julie Bouckaert
- UGSF–UMR8576, University of Lille, CNRS, F-59000 Lille, France
| | | | - Xavier Hanoulle
- UGSF–UMR8576, University of Lille, CNRS, F-59000 Lille, France
| |
Collapse
|
30
|
Mackman RL, Steadman VA, Dean DK, Jansa P, Poullennec KG, Appleby T, Austin C, Blakemore CA, Cai R, Cannizzaro C, Chin G, Chiva JYC, Dunbar NA, Fliri H, Highton AJ, Hui H, Ji M, Jin H, Karki K, Keats AJ, Lazarides L, Lee YJ, Liclican A, Mish M, Murray B, Pettit SB, Pyun P, Sangi M, Santos R, Sanvoisin J, Schmitz U, Schrier A, Siegel D, Sperandio D, Stepan G, Tian Y, Watt GM, Yang H, Schultz BE. Discovery of a Potent and Orally Bioavailable Cyclophilin Inhibitor Derived from the Sanglifehrin Macrocycle. J Med Chem 2018; 61:9473-9499. [PMID: 30074795 DOI: 10.1021/acs.jmedchem.8b00802] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Cyclophilins are a family of peptidyl-prolyl isomerases that are implicated in a wide range of diseases including hepatitis C. Our aim was to discover through total synthesis an orally bioavailable, non-immunosuppressive cyclophilin (Cyp) inhibitor with potent anti-hepatitis C virus (HCV) activity that could serve as part of an all oral antiviral combination therapy. An initial lead 2 derived from the sanglifehrin A macrocycle was optimized using structure based design to produce a potent and orally bioavailable inhibitor 3. The macrocycle ring size was reduced by one atom, and an internal hydrogen bond drove improved permeability and drug-like properties. 3 demonstrates potent Cyp inhibition ( Kd = 5 nM), potent anti-HCV 2a activity (EC50 = 98 nM), and high oral bioavailability in rat (100%) and dog (55%). The synthetic accessibility and properties of 3 support its potential as an anti-HCV agent and for interrogating the role of Cyp inhibition in a variety of diseases.
Collapse
Affiliation(s)
- Richard L Mackman
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Victoria A Steadman
- Selcia Ltd. , Fyfield Business and Research Park, Fyfield Road , Ongar , Essex CM5 0GS , United Kingdom
| | - David K Dean
- Selcia Ltd. , Fyfield Business and Research Park, Fyfield Road , Ongar , Essex CM5 0GS , United Kingdom
| | - Petr Jansa
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Karine G Poullennec
- Selcia Ltd. , Fyfield Business and Research Park, Fyfield Road , Ongar , Essex CM5 0GS , United Kingdom
| | - Todd Appleby
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Carol Austin
- Selcia Ltd. , Fyfield Business and Research Park, Fyfield Road , Ongar , Essex CM5 0GS , United Kingdom
| | - Caroline A Blakemore
- Selcia Ltd. , Fyfield Business and Research Park, Fyfield Road , Ongar , Essex CM5 0GS , United Kingdom
| | - Ruby Cai
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Carina Cannizzaro
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Gregory Chin
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Jean-Yves C Chiva
- Selcia Ltd. , Fyfield Business and Research Park, Fyfield Road , Ongar , Essex CM5 0GS , United Kingdom
| | - Neil A Dunbar
- Selcia Ltd. , Fyfield Business and Research Park, Fyfield Road , Ongar , Essex CM5 0GS , United Kingdom
| | - Hans Fliri
- Cypralis Ltd. , Babraham Research Campus, Cambridge CB22 3AT , United Kingdom
| | - Adrian J Highton
- Selcia Ltd. , Fyfield Business and Research Park, Fyfield Road , Ongar , Essex CM5 0GS , United Kingdom
| | - Hon Hui
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Mingzhe Ji
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Haolun Jin
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Kapil Karki
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Andrew J Keats
- Selcia Ltd. , Fyfield Business and Research Park, Fyfield Road , Ongar , Essex CM5 0GS , United Kingdom
| | - Linos Lazarides
- Selcia Ltd. , Fyfield Business and Research Park, Fyfield Road , Ongar , Essex CM5 0GS , United Kingdom
| | - Yu-Jen Lee
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Albert Liclican
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Michael Mish
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Bernard Murray
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Simon B Pettit
- Selcia Ltd. , Fyfield Business and Research Park, Fyfield Road , Ongar , Essex CM5 0GS , United Kingdom
| | - Peter Pyun
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Michael Sangi
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Rex Santos
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Jonathan Sanvoisin
- Selcia Ltd. , Fyfield Business and Research Park, Fyfield Road , Ongar , Essex CM5 0GS , United Kingdom
| | - Uli Schmitz
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Adam Schrier
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Dustin Siegel
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - David Sperandio
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - George Stepan
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Yang Tian
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Gregory M Watt
- Selcia Ltd. , Fyfield Business and Research Park, Fyfield Road , Ongar , Essex CM5 0GS , United Kingdom
| | - Hai Yang
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| | - Brian E Schultz
- Gilead Sciences Inc. , 333 Lakeside Drive , Foster City , California 94404 , United States
| |
Collapse
|
31
|
Fu J, Becker C, Cao L, Capparelli M, Denay R, Fujimoto R, Gai Y, Gao Z, Guenat C, Karur S, Kim H, Li W, Li X, Li W, Lochmann T, Lu A, Lu P, Luneau A, Meier N, Mergo W, Ng S, Parker D, Peng Y, Riss B, Rivkin A, Roggo S, Schroeder H, Schuerch F, Simmons RL, Sun F, Sweeney ZK, Tjandra M, Wang M, Wang R, Weiss AH, Wenger N, Wu Q, Xiong X, Xu S, Xu W, Yifru A, Zhao J, Zhou J, Zürcher C, Gallou F. Development of a cyclosporin A derivative with excellent anti-hepatitis C virus potency. Bioorg Med Chem 2018; 26:957-969. [DOI: 10.1016/j.bmc.2017.09.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 08/21/2017] [Accepted: 09/06/2017] [Indexed: 12/27/2022]
|
32
|
Kahlert V, Prell E, Ohlenschläger O, Melesina J, Schumann M, Lücke C, Fischer G, Malešević M. Synthesis and biochemical evaluation of two novel N-hydroxyalkylated cyclosporin A analogs. Org Biomol Chem 2018; 16:4338-4349. [DOI: 10.1039/c8ob00980e] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
N-Hydroxyalkylation of cyclosporine A residues Val5 and d-Ala8 significantly influenced their conformation behavior and pharmacological properties.
Collapse
Affiliation(s)
- Viktoria Kahlert
- Max Planck Research Unit for Enzymology of Protein Folding
- 06120 Halle/Saale
- Germany
| | - Erik Prell
- Max Planck Research Unit for Enzymology of Protein Folding
- 06120 Halle/Saale
- Germany
- Max-Planck Institute for Biophysical Chemistry
- Göttingen
| | | | - Jelena Melesina
- Martin-Luther-University Halle-Wittenberg
- Institute of Pharmacy
- Medicinal Chemistry department
- 06120 Halle
- Germany
| | - Michael Schumann
- Max Planck Research Unit for Enzymology of Protein Folding
- 06120 Halle/Saale
- Germany
- Martin-Luther-University Halle-Wittenberg
- Institute of Biochemistry and Biotechnology
| | - Christian Lücke
- Max Planck Research Unit for Enzymology of Protein Folding
- 06120 Halle/Saale
- Germany
| | - Gunter Fischer
- Max Planck Research Unit for Enzymology of Protein Folding
- 06120 Halle/Saale
- Germany
- Max-Planck Institute for Biophysical Chemistry
- Göttingen
| | - Miroslav Malešević
- Max Planck Research Unit for Enzymology of Protein Folding
- 06120 Halle/Saale
- Germany
- Martin-Luther-University Halle-Wittenberg
- Institute of Biochemistry and Biotechnology
| |
Collapse
|
33
|
Liu H, Huang D, Jin L, Wang C, Liang S, Wen J. Integrating multi-omics analyses of Nonomuraea dietziae to reveal the role of soybean oil in [(4'-OH)MeLeu] 4-CsA overproduction. Microb Cell Fact 2017; 16:120. [PMID: 28709434 PMCID: PMC5512743 DOI: 10.1186/s12934-017-0739-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 07/10/2017] [Indexed: 12/29/2022] Open
Abstract
Background Nonomuraea dietziae is a promising microorganism to mediate the region-specific monooxygenation reaction of cyclosporine A (CsA). The main product [(4′-OH)MeLeu]4-CsA possesses high anti-HIV/HCV and hair growth-stimulating activities while avoiding the immunosuppressive effect of CsA. However, the low conversion efficiency restricts the clinical application. In this study, the production of [(4′-OH)MeLeu]4-CsA was greatly improved by 55.6% from 182.8 to 284.4 mg/L when supplementing soybean oil into the production medium, which represented the highest production of [(4′-OH)MeLeu]4-CsA so far. Results To investigate the effect of soybean oil on CsA conversion, some other plant oils (corn oil and peanut oil) and the major hydrolysates of soybean oil were fed into the production medium, respectively. The results demonstrated that the plant oils, rather than the hydrolysates, could significantly improve the [(4′-OH)MeLeu]4-CsA production, suggesting that soybean oil might not play its role in the lipid metabolic pathway. To further unveil the mechanism of [(4′-OH)MeLeu]4-CsA overproduction under the soybean oil condition, a proteomic analysis based on the two-dimensional gel electrophoresis coupled with MALDI TOF/TOF mass spectrometry was implemented. The results showed that central carbon metabolism, genetic information processing and energy metabolism were significantly up-regulated under the soybean oil condition. Moreover, the gas chromatography-mass spectrometry-based metabolomic analysis indicated that soybean oil had a great effect on amino acid metabolism and tricarboxylic acid cycle. In addition, the transcription levels of cytochrome P450 hydroxylase (CYP) genes for CsA conversion were determined by RT-qPCR and the results showed that most of the CYP genes were up-regulated under the soybean oil condition. Conclusions These findings indicate that soybean oil could strengthen the primary metabolism and the CYP system to enhance the mycelium growth and the monooxygenation reaction, respectively, and it will be a guidance for the further metabolic engineering of this strain. Electronic supplementary material The online version of this article (doi:10.1186/s12934-017-0739-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Huanhuan Liu
- Key Laboratory of System Bioengineering (Tianjin University), Ministry of Education, Tianjin, 300072, People's Republic of China.,SynBio Research Platform, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, People's Republic of China
| | - Di Huang
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin, 300457, People's Republic of China.,SynBio Research Platform, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin, 300071, People's Republic of China
| | - Lina Jin
- Key Laboratory of System Bioengineering (Tianjin University), Ministry of Education, Tianjin, 300072, People's Republic of China.,SynBio Research Platform, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, People's Republic of China
| | - Cheng Wang
- Key Laboratory of System Bioengineering (Tianjin University), Ministry of Education, Tianjin, 300072, People's Republic of China.,SynBio Research Platform, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, People's Republic of China
| | - Shaoxiong Liang
- Key Laboratory of System Bioengineering (Tianjin University), Ministry of Education, Tianjin, 300072, People's Republic of China.,SynBio Research Platform, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, People's Republic of China
| | - Jianping Wen
- Key Laboratory of System Bioengineering (Tianjin University), Ministry of Education, Tianjin, 300072, People's Republic of China. .,SynBio Research Platform, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, People's Republic of China.
| |
Collapse
|
34
|
Price DA, Eng H, Farley KA, Goetz GH, Huang Y, Jiao Z, Kalgutkar AS, Kablaoui NM, Khunte B, Liras S, Limberakis C, Mathiowetz AM, Ruggeri RB, Quan JM, Yang Z. Comparative pharmacokinetic profile of cyclosporine (CsA) with a decapeptide and a linear analogue. Org Biomol Chem 2017; 15:2501-2506. [PMID: 28266673 DOI: 10.1039/c7ob00096k] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The synthesis and in vivo pharmacokinetic profile of an analogue of cyclosporine is disclosed. An acyclic congener was also profiled in in vitro assays to compare cell permeability. The compounds possess similar calculated and measured molecular descriptors however have different behaviors in an RRCK assay to assess cell permeability.
Collapse
Affiliation(s)
- David A Price
- Pfizer Inc. Medicine Design, 610 Main Street, Cambridge, Massachusetts, 02155 USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Juvvadi PR, Lee SC, Heitman J, Steinbach WJ. Calcineurin in fungal virulence and drug resistance: Prospects for harnessing targeted inhibition of calcineurin for an antifungal therapeutic approach. Virulence 2017; 8:186-197. [PMID: 27325145 PMCID: PMC5354160 DOI: 10.1080/21505594.2016.1201250] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 01/26/2023] Open
Abstract
Increases in the incidence and mortality due to the major invasive fungal infections such as aspergillosis, candidiasis and cryptococcosis caused by the species of Aspergillus, Candida and Cryptococcus, are a growing threat to the immunosuppressed patient population. In addition to the limited armamentarium of the current classes of antifungal agents available (pyrimidine analogs, polyenes, azoles, and echinocandins), their toxicity, efficacy and the emergence of resistance are major bottlenecks limiting successful patient outcomes. Although these drugs target distinct fungal pathways, there is an urgent need to develop new antifungals that are more efficacious, fungal-specific, with reduced or no toxicity and simultaneously do not induce resistance. Here we review several lines of evidence which indicate that the calcineurin signaling pathway, a target of the immunosuppressive drugs FK506 and cyclosporine A, orchestrates growth, virulence and drug resistance in a variety of fungal pathogens and can be exploited for novel antifungal drug development.
Collapse
Affiliation(s)
- Praveen R. Juvvadi
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Soo Chan Lee
- Department of Molecular Genetics & Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Joseph Heitman
- Department of Molecular Genetics & Microbiology, Duke University School of Medicine, Durham, NC, USA
- Department of Medicine, Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - William J. Steinbach
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics & Microbiology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
36
|
Steadman VA, Pettit SB, Poullennec KG, Lazarides L, Keats AJ, Dean DK, Stanway SJ, Austin CA, Sanvoisin JA, Watt GM, Fliri HG, Liclican AC, Jin D, Wong MH, Leavitt SA, Lee YJ, Tian Y, Frey CR, Appleby TC, Schmitz U, Jansa P, Mackman RL, Schultz BE. Discovery of Potent Cyclophilin Inhibitors Based on the Structural Simplification of Sanglifehrin A. J Med Chem 2017; 60:1000-1017. [DOI: 10.1021/acs.jmedchem.6b01329] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Victoria A. Steadman
- Selcia Ltd., Fyfield Business & Research Park, Fyfield Road, Ongar, Essex CM5 0GS, United Kingdom
| | - Simon B. Pettit
- Selcia Ltd., Fyfield Business & Research Park, Fyfield Road, Ongar, Essex CM5 0GS, United Kingdom
| | - Karine G. Poullennec
- Selcia Ltd., Fyfield Business & Research Park, Fyfield Road, Ongar, Essex CM5 0GS, United Kingdom
| | - Linos Lazarides
- Selcia Ltd., Fyfield Business & Research Park, Fyfield Road, Ongar, Essex CM5 0GS, United Kingdom
| | - Andrew J. Keats
- Selcia Ltd., Fyfield Business & Research Park, Fyfield Road, Ongar, Essex CM5 0GS, United Kingdom
| | - David K. Dean
- Selcia Ltd., Fyfield Business & Research Park, Fyfield Road, Ongar, Essex CM5 0GS, United Kingdom
| | - Steven J. Stanway
- Selcia Ltd., Fyfield Business & Research Park, Fyfield Road, Ongar, Essex CM5 0GS, United Kingdom
| | - Carol A. Austin
- Selcia Ltd., Fyfield Business & Research Park, Fyfield Road, Ongar, Essex CM5 0GS, United Kingdom
| | - Jonathan A. Sanvoisin
- Selcia Ltd., Fyfield Business & Research Park, Fyfield Road, Ongar, Essex CM5 0GS, United Kingdom
| | - Gregory M. Watt
- Selcia Ltd., Fyfield Business & Research Park, Fyfield Road, Ongar, Essex CM5 0GS, United Kingdom
| | - Hans G. Fliri
- Cypralis Ltd., Babraham Research
Campus, Cambridge CB22
3AT, United Kingdom
| | - Albert C. Liclican
- Gilead Sciences, 333 Lakeside
Drive, Foster City, California 94404, United States
| | - Debi Jin
- Gilead Sciences, 333 Lakeside
Drive, Foster City, California 94404, United States
| | - Melanie H. Wong
- Gilead Sciences, 333 Lakeside
Drive, Foster City, California 94404, United States
| | - Stephanie A. Leavitt
- Gilead Sciences, 333 Lakeside
Drive, Foster City, California 94404, United States
| | - Yu-Jen Lee
- Gilead Sciences, 333 Lakeside
Drive, Foster City, California 94404, United States
| | - Yang Tian
- Gilead Sciences, 333 Lakeside
Drive, Foster City, California 94404, United States
| | - Christian R. Frey
- Gilead Sciences, 333 Lakeside
Drive, Foster City, California 94404, United States
| | - Todd C. Appleby
- Gilead Sciences, 333 Lakeside
Drive, Foster City, California 94404, United States
| | - Uli Schmitz
- Gilead Sciences, 333 Lakeside
Drive, Foster City, California 94404, United States
| | - Petr Jansa
- Gilead Sciences, 333 Lakeside
Drive, Foster City, California 94404, United States
| | - Richard L. Mackman
- Gilead Sciences, 333 Lakeside
Drive, Foster City, California 94404, United States
| | - Brian E. Schultz
- Gilead Sciences, 333 Lakeside
Drive, Foster City, California 94404, United States
| |
Collapse
|
37
|
Ahmed-Belkacem A, Colliandre L, Ahnou N, Nevers Q, Gelin M, Bessin Y, Brillet R, Cala O, Douguet D, Bourguet W, Krimm I, Pawlotsky JM, Guichou JF. Fragment-based discovery of a new family of non-peptidic small-molecule cyclophilin inhibitors with potent antiviral activities. Nat Commun 2016; 7:12777. [PMID: 27652979 PMCID: PMC5036131 DOI: 10.1038/ncomms12777] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 08/01/2016] [Indexed: 12/16/2022] Open
Abstract
Cyclophilins are peptidyl-prolyl cis/trans isomerases (PPIase) that catalyse the interconversion of the peptide bond at proline residues. Several cyclophilins play a pivotal role in the life cycle of a number of viruses. The existing cyclophilin inhibitors, all derived from cyclosporine A or sanglifehrin A, have disadvantages, including their size, potential for side effects unrelated to cyclophilin inhibition and drug–drug interactions, unclear antiviral spectrum and manufacturing issues. Here we use a fragment-based drug discovery approach using nucleic magnetic resonance, X-ray crystallography and structure-based compound optimization to generate a new family of non-peptidic, small-molecule cyclophilin inhibitors with potent in vitro PPIase inhibitory activity and antiviral activity against hepatitis C virus, human immunodeficiency virus and coronaviruses. This family of compounds has the potential for broad-spectrum, high-barrier-to-resistance treatment of viral infections. Cyclophilins play a key role in the life cycle of many viruses and represent important drug targets for broad-spectrum antiviral therapies. Here, the authors use fragment-based drug discovery to develop non-peptidic inhibitors of human cyclophilins with high activity against replication of a number of viral families.
Collapse
Affiliation(s)
- Abdelhakim Ahmed-Belkacem
- INSERM U955 'Pathophysiology and Therapy of Chronic Viral Hepatitis and Related Cancers', Hôpital Henri Mondor, Université Paris-Est, 51 avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Lionel Colliandre
- CNRS UMR5048, Centre de Biochimie Structurale, Université de Montpellier, 29 rue de Navacelles, 34090 Montpellier, France.,INSERM U1054, Centre de Biochimie Structurale, Université de Montpellier, 29 rue de Navacelles, 34090 Montpellier, France
| | - Nazim Ahnou
- INSERM U955 'Pathophysiology and Therapy of Chronic Viral Hepatitis and Related Cancers', Hôpital Henri Mondor, Université Paris-Est, 51 avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Quentin Nevers
- INSERM U955 'Pathophysiology and Therapy of Chronic Viral Hepatitis and Related Cancers', Hôpital Henri Mondor, Université Paris-Est, 51 avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Muriel Gelin
- CNRS UMR5048, Centre de Biochimie Structurale, Université de Montpellier, 29 rue de Navacelles, 34090 Montpellier, France.,INSERM U1054, Centre de Biochimie Structurale, Université de Montpellier, 29 rue de Navacelles, 34090 Montpellier, France
| | - Yannick Bessin
- CNRS UMR5048, Centre de Biochimie Structurale, Université de Montpellier, 29 rue de Navacelles, 34090 Montpellier, France.,INSERM U1054, Centre de Biochimie Structurale, Université de Montpellier, 29 rue de Navacelles, 34090 Montpellier, France
| | - Rozenn Brillet
- INSERM U955 'Pathophysiology and Therapy of Chronic Viral Hepatitis and Related Cancers', Hôpital Henri Mondor, Université Paris-Est, 51 avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Olivier Cala
- Institut des Sciences Analytiques, CNRS UMR5280, Université Lyon 1, École Nationale Supérieure de Lyon, 5 rue de la Doua, 69100 Villeurbanne, France
| | - Dominique Douguet
- CNRS UMR5048, Centre de Biochimie Structurale, Université de Montpellier, 29 rue de Navacelles, 34090 Montpellier, France.,INSERM U1054, Centre de Biochimie Structurale, Université de Montpellier, 29 rue de Navacelles, 34090 Montpellier, France
| | - William Bourguet
- CNRS UMR5048, Centre de Biochimie Structurale, Université de Montpellier, 29 rue de Navacelles, 34090 Montpellier, France.,INSERM U1054, Centre de Biochimie Structurale, Université de Montpellier, 29 rue de Navacelles, 34090 Montpellier, France
| | - Isabelle Krimm
- Institut des Sciences Analytiques, CNRS UMR5280, Université Lyon 1, École Nationale Supérieure de Lyon, 5 rue de la Doua, 69100 Villeurbanne, France
| | - Jean-Michel Pawlotsky
- INSERM U955 'Pathophysiology and Therapy of Chronic Viral Hepatitis and Related Cancers', Hôpital Henri Mondor, Université Paris-Est, 51 avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France.,National Reference Center for Viral Hepatitis B, C and Delta, Department of Virology, Hôpital Henri Mondor, Université Paris-Est, 51 avenue du Maréchal de Lattre de Tassigny, 94010 Créteil, France
| | - Jean-François Guichou
- CNRS UMR5048, Centre de Biochimie Structurale, Université de Montpellier, 29 rue de Navacelles, 34090 Montpellier, France.,INSERM U1054, Centre de Biochimie Structurale, Université de Montpellier, 29 rue de Navacelles, 34090 Montpellier, France
| |
Collapse
|
38
|
Chiramel AI, Banadyga L, Dougherty JD, Falzarano D, Martellaro C, Brees D, Taylor RT, Ebihara H, Best SM. Alisporivir Has Limited Antiviral Effects Against Ebola Virus Strains Makona and Mayinga. J Infect Dis 2016; 214:S355-S359. [PMID: 27511894 DOI: 10.1093/infdis/jiw241] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Antiviral therapeutics with existing clinical safety profiles would be highly desirable in an outbreak situation, such as the 2013-2016 emergence of Ebola virus (EBOV) in West Africa. Although, the World Health Organization declared the end of the outbreak early 2016, sporadic cases of EBOV infection have since been reported. Alisporivir is the most clinically advanced broad-spectrum antiviral that functions by targeting a host protein, cyclophilin A (CypA). A modest antiviral effect of alisporivir against contemporary (Makona) but not historical (Mayinga) EBOV strains was observed in tissue culture. However, this effect was not comparable to observations for an alisporivir-susceptible virus, the flavivirus tick-borne encephalitis virus. Thus, EBOV does not depend on (CypA) for replication, in contrast to many other viruses pathogenic to humans.
Collapse
Affiliation(s)
- Abhilash I Chiramel
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana
| | - Logan Banadyga
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana
| | - Jonathan D Dougherty
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana
| | - Darryl Falzarano
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana Vaccine and Infectious Disease Organization-International Vaccine Centre, University of Saskatchewan, Saskatoon, Canada
| | - Cynthia Martellaro
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana
| | | | - R Travis Taylor
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana Department of Medical Microbiology and Immunology, College of Medicine, University of Toledo Health Science Campus, Ohio
| | - Hideki Ebihara
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana
| | - Sonja M Best
- Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, Montana
| |
Collapse
|
39
|
Ma C, Li F, Musharrafieh RG, Wang J. Discovery of cyclosporine A and its analogs as broad-spectrum anti-influenza drugs with a high in vitro genetic barrier of drug resistance. Antiviral Res 2016; 133:62-72. [PMID: 27478032 DOI: 10.1016/j.antiviral.2016.07.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 07/26/2016] [Indexed: 12/13/2022]
Abstract
As the number of drug-resistant influenza viruses continues to increase, antivirals with novel mechanisms of action are urgently needed. Among the two classes of FDA-approved antiviral drugs, neuraminidase (NA) inhibitors, oseltamivir, zanamivir, and peramivir, are currently the only choice for the prevention and treatment of influenza virus infection. Due to the antigenic drift and antigenic shift, it will only be a matter of time before influenza viruses become completely resistant to these NA inhibitors. In pursuing the next generation of antiviral drugs with complementary mechanisms of action to those of the NA inhibitors, we have identified a natural product, cyclosporine A (CsA) (1), as a desired drug candidate. In this study, we discovered that CsA (1) and its analogs have broad-spectrum antiviral activity against multiple influenza A and B strains, including strains that are resistant to either NA or M2 inhibitors or both. Moreover, CsA (1) displays a high in vitro genetic barrier of drug resistance than oseltamivir carboxylate Mechanistic studies revealed that CsA (1) acts at the intermediate step of viral replication post viral fusion. Its antiviral mechanism is independent of inhibiting the isomerase activity of cyclophilin A (CypA), and CsA (1) has no effect on the viral polymerase activity The potent antiviral efficacy of CsA (1), coupled with the high in vitro genetic barrier of drug resistance and novel mechanism of action, renders CsA (1) a promising anti-influenza drug candidate for further development.
Collapse
Affiliation(s)
- Chunlong Ma
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States; BIO5 Institute, The University of Arizona, Tucson, AZ 85721, United States
| | - Fang Li
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States; BIO5 Institute, The University of Arizona, Tucson, AZ 85721, United States
| | - Rami Ghassan Musharrafieh
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ 85721, United States
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, United States; BIO5 Institute, The University of Arizona, Tucson, AZ 85721, United States.
| |
Collapse
|
40
|
Joshi AU, Kornfeld OS, Mochly-Rosen D. The entangled ER-mitochondrial axis as a potential therapeutic strategy in neurodegeneration: A tangled duo unchained. Cell Calcium 2016; 60:218-34. [PMID: 27212603 DOI: 10.1016/j.ceca.2016.04.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 04/28/2016] [Accepted: 04/28/2016] [Indexed: 12/12/2022]
Abstract
Endoplasmic reticulum (ER) and mitochondrial function have both been shown to be critical events in neurodegenerative diseases. The ER mediates protein folding, maturation, sorting as well acts as calcium storage. The unfolded protein response (UPR) is a stress response of the ER that is activated by the accumulation of misfolded proteins within the ER lumen. Although the molecular mechanisms underlying ER stress-induced apoptosis are not completely understood, increasing evidence suggests that ER and mitochondria cooperate to signal cell death. Similarly, calcium-mediated mitochondrial function and dynamics not only contribute to ATP generation and calcium buffering but are also a linchpin in mediating cell fate. Mitochondria and ER form structural and functional networks (mitochondria-associated ER membranes [MAMs]) essential to maintaining cellular homeostasis and determining cell fate under various pathophysiological conditions. Regulated Ca(2+) transfer from the ER to the mitochondria is important in maintaining control of pro-survival/pro-death pathways. In this review, we summarize the latest therapeutic strategies that target these essential organelles in the context of neurodegenerative diseases.
Collapse
Affiliation(s)
- Amit U Joshi
- Department of Chemical & Systems Biology, School of Medicine, Stanford University, CA, USA
| | - Opher S Kornfeld
- Department of Chemical & Systems Biology, School of Medicine, Stanford University, CA, USA
| | - Daria Mochly-Rosen
- Department of Chemical & Systems Biology, School of Medicine, Stanford University, CA, USA.
| |
Collapse
|
41
|
Gallay PA, Bobardt MD, Chatterji U, Trepanier DJ, Ure D, Ordonez C, Foster R. The Novel Cyclophilin Inhibitor CPI-431-32 Concurrently Blocks HCV and HIV-1 Infections via a Similar Mechanism of Action. PLoS One 2015; 10:e0134707. [PMID: 26263487 PMCID: PMC4532424 DOI: 10.1371/journal.pone.0134707] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 07/13/2015] [Indexed: 12/17/2022] Open
Abstract
HCV-related liver disease is the main cause of morbidity and mortality of HCV/HIV-1 co-infected patients. Despite the recent advent of anti-HCV direct acting antivirals (DAAs), the treatment of HCV/HIV-1 co-infected patients remains a challenge, as these patients are refractory to most therapies and develop liver fibrosis, cirrhosis and liver cancer more often than HCV mono-infected patients. Until the present study, there was no suitable in vitro assay to test the inhibitory activity of drugs on HCV/HIV-1 co-infection. Here we developed a novel in vitro "co-infection" model where HCV and HIV-1 concurrently replicate in their respective main host target cells--human hepatocytes and CD4+ T-lymphocytes. Using this co-culture model, we demonstrate that cyclophilin inhibitors (CypI), including a novel cyclosporin A (CsA) analog, CPI-431-32, simultaneously inhibits replication of both HCV and HIV-1 when added pre- and post-infection. In contrast, the HIV-1 protease inhibitor nelfinavir or the HCV NS5A inhibitor daclatasvir only blocks the replication of a single virus in the "co-infection" system. CPI-431-32 efficiently inhibits HCV and HIV-1 variants, which are normally resistant to DAAs. CPI-431-32 is slightly, but consistently more efficacious than the most advanced clinically tested CypI--alisporivir (ALV)--at interrupting an established HCV/HIV-1 co-infection. The superior antiviral efficacy of CPI-431-32 over ALV correlates with its higher potency inhibition of cyclophilin A (CypA) isomerase activity and at preventing HCV NS5A-CypA and HIV-1 capsid-CypA interactions known to be vital for replication of the respective viruses. Moreover, we obtained evidence that CPI-431-32 prevents the cloaking of both the HIV-1 and HCV genomes from cellular sensors. Based on these results, CPI-431-32 has the potential, as a single agent or in combination with DAAs, to inhibit both HCV and HIV-1 infections.
Collapse
Affiliation(s)
- Philippe A. Gallay
- Department of Immunology & Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Michael D. Bobardt
- Department of Immunology & Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Udayan Chatterji
- Department of Immunology & Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Daniel J. Trepanier
- Ciclofilin Pharmaceuticals Inc., San Diego, California, United States of America
| | - Daren Ure
- Ciclofilin Pharmaceuticals Inc., San Diego, California, United States of America
| | - Cosme Ordonez
- Ciclofilin Pharmaceuticals Inc., San Diego, California, United States of America
| | - Robert Foster
- Ciclofilin Pharmaceuticals Inc., San Diego, California, United States of America
| |
Collapse
|
42
|
Kornfeld OS, Hwang S, Disatnik MH, Chen CH, Qvit N, Mochly-Rosen D. Mitochondrial reactive oxygen species at the heart of the matter: new therapeutic approaches for cardiovascular diseases. Circ Res 2015; 116:1783-99. [PMID: 25999419 DOI: 10.1161/circresaha.116.305432] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Reactive oxygen species (ROS) have been implicated in a variety of age-related diseases, including multiple cardiovascular disorders. However, translation of ROS scavengers (antioxidants) into the clinic has not been successful. These antioxidants grossly reduce total levels of cellular ROS including ROS that participate in physiological signaling. In this review, we challenge the traditional antioxidant therapeutic approach that targets ROS directly with novel approaches that improve mitochondrial functions to more effectively treat cardiovascular diseases.
Collapse
Affiliation(s)
- Opher S Kornfeld
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA
| | - Sunhee Hwang
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA
| | - Marie-Hélène Disatnik
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA
| | - Che-Hong Chen
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA
| | - Nir Qvit
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA
| | - Daria Mochly-Rosen
- From the Department of Chemical and Systems Biology, Stanford University School of Medicine, CA.
| |
Collapse
|
43
|
Esser‐Nobis K, Harak C, Schult P, Kusov Y, Lohmann V. Novel perspectives for hepatitis A virus therapy revealed by comparative analysis of hepatitis C virus and hepatitis A virus RNA replication. Hepatology 2015; 62:397-408. [PMID: 25866017 PMCID: PMC7165973 DOI: 10.1002/hep.27847] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 04/10/2015] [Indexed: 12/11/2022]
Abstract
UNLABELLED Hepatitis A virus (HAV) and hepatitis C virus (HCV) are two positive-strand RNA viruses sharing a similar biology, but causing opposing infection outcomes, with HAV always being cleared and HCV establishing persistence in the majority of infections. To gain deeper insight into determinants of replication, persistence, and treatment, we established a homogenous cell-culture model allowing a thorough comparison of RNA replication of both viruses. By screening different human liver-derived cell lines with subgenomic reporter replicons of HAV as well as of different HCV genotypes, we found that Huh7-Lunet cells supported HAV- and HCV-RNA replication with similar efficiency and limited interference between both replicases. HAV and HCV replicons were similarly sensitive to interferon (IFN), but differed in their ability to establish persistent replication in cell culture. In contrast to HCV, HAV replicated independently from microRNA-122 and phosphatidylinositol 4-kinase IIIα and β (PI4KIII). Both viruses were efficiently inhibited by cyclosporin A and NIM811, a nonimmunosuppressive analog thereof, suggesting an overlapping dependency on cyclophilins for replication. However, analysis of a broader set of inhibitors revealed that, in contrast to HCV, HAV does not depend on cyclophilin A, but rather on adenosine-triphosphate-binding cassette transporters and FK506-binding proteins. Finally, silibinin, but not its modified intravenous formulation, efficiently inhibited HAV genome replication in vitro, suggesting oral silibinin as a potential therapeutic option for HAV infections. CONCLUSION We established a cell-culture model enabling comparative studies on RNA replication of HAV and HCV in a homogenous cellular background with comparable replication efficiency. We thereby identified new host cell targets and potential treatment options for HAV and set the ground for future studies to unravel determinants of clearance and persistence.
Collapse
Affiliation(s)
- Katharina Esser‐Nobis
- Department of Infectious DiseasesMolecular Virology, University of HeidelbergGermany
| | - Christian Harak
- Department of Infectious DiseasesMolecular Virology, University of HeidelbergGermany
| | - Philipp Schult
- Department of Infectious DiseasesMolecular Virology, University of HeidelbergGermany
| | - Yuri Kusov
- Institute of Biochemistry, Center for Structural and Cell Biology in Medicine and German Center for Infection Research (DZIF), University of LübeckGermany
| | - Volker Lohmann
- Department of Infectious DiseasesMolecular Virology, University of HeidelbergGermany
| |
Collapse
|
44
|
Semi-synthesis of cyclosporins. Biochim Biophys Acta Gen Subj 2015; 1850:2121-44. [PMID: 25707381 DOI: 10.1016/j.bbagen.2015.02.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 02/11/2015] [Accepted: 02/13/2015] [Indexed: 12/28/2022]
Abstract
BACKGROUND Since its isolation in 1970, and discovery of its potent inhibitory activity on T-cell proliferation, cyclosporin A (CsA) has been shown to play a significant role in diverse fields of biology. Furthermore, chemical modification of CsA has led to analogs with distinct biological activities associated with its protein receptor family, cyclophilins. SCOPE OF REVIEW This review systematically collates the synthetic chemistry performed at each of the eleven amino acids, and provides examples of the utility of such transformations. The various modifications of CsA are traced from early, modest chemistry performed at the unique Bmt residue, through the remarkable use of a polyanion enolate that can be stereoselectively manipulated, and onto application of more recently developed olefin metathesis chemistry to prepare new CsA derivatives with unexpected biological activity. MAJOR CONCLUSIONS The myriad biological activities of CsA and its synthetic derivatives have inspired the development of new approaches to modify the CsA ring. In turn, these new CsA derivatives have served as tools in the discovery of new roles for cyclophilins. GENERAL SIGNIFICANCE This review provides information on the types of cyclosporin derivatives that are available to the many biologists working in this field, and should be of value to the medicinal chemist trying to discover drugs based on CsA. This article is part of a Special Issue entitled Proline-directed foldases: Cell signaling catalysts and drug targets.
Collapse
|
45
|
Zhu DY, Zhang Z, Mou XQ, Tu YQ, Zhang FM, Peng JB, Wang SH, Zhang SY. Gold(I)/Copper(II)-Cocatalyzed Tandem Cyclization/Semipinacol Reaction: Construction of 6-Aza/Oxa-Spiro[4.5]decane Skeletons and Formal Synthesis of (±)-Halichlorine. Adv Synth Catal 2015. [DOI: 10.1002/adsc.201400932] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
46
|
Lawen A. Biosynthesis of cyclosporins and other natural peptidyl prolyl cis/trans isomerase inhibitors. Biochim Biophys Acta Gen Subj 2014; 1850:2111-20. [PMID: 25497210 DOI: 10.1016/j.bbagen.2014.12.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 12/01/2014] [Accepted: 12/04/2014] [Indexed: 01/11/2023]
Abstract
BACKGROUND Peptidyl-prolyl-cis/trans-isomerases (PPIases) are ubiquitously expressed and have been implicated in a wide range of biological functions. Their inhibition is beneficial in immunosuppression, cancer treatment, treatment of autoimmune diseases, protozoan and viral infections. SCOPE OF REVIEW Three classes of PPIases are known, each class having their own specific inhibitors. This review will cover the present knowledge on the biosynthesis of the natural PPIase inhibitors. These include for the cyclophilins: the cyclosporins, the analogues of peptolide SDZ 214-103 and the sanglifehrins; for the FKBPs: ascomycin, rapamycin and FK506 and for the parvulins the naphtoquinone juglone. MAJOR CONCLUSIONS Over the last thirty years much progress has been made in understanding PPIase function and the biosynthesis of natural PPIase inhibitors. Non-immunosuppressive analogues were discovered and served as lead compounds for the development of novel antiviral drugs. There are, however, still unsolved questions which deserve further research into this exciting field. GENERAL SIGNIFICANCE As all the major natural inhibitors of the cyclophilins and FKBPs are synthesized by complex non-ribosomal peptide synthetases and/or polyketide synthases, total chemical synthesis is not a viable option. Thus, fully understanding the modular enzyme systems involved in their biosynthesis may help engineering enzymes capable of synthesizing novel PPIase inhibitors with improved functions for a wide range of conditions. This article is part of a Special Issue entitled Proline-directed Foldases: Cell signaling catalysts and drug targets.
Collapse
Affiliation(s)
- Alfons Lawen
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Victoria 3800, Australia.
| |
Collapse
|
47
|
Fu J, Tjandra M, Becker C, Bednarczyk D, Capparelli M, Elling R, Hanna I, Fujimoto R, Furegati M, Karur S, Kasprzyk T, Knapp M, Leung K, Li X, Lu P, Mergo W, Miault C, Ng S, Parker D, Peng Y, Roggo S, Rivkin A, Simmons RL, Wang M, Wiedmann B, Weiss AH, Xiao L, Xie L, Xu W, Yifru A, Yang S, Zhou B, Sweeney ZK. Potent nonimmunosuppressive cyclophilin inhibitors with improved pharmaceutical properties and decreased transporter inhibition. J Med Chem 2014; 57:8503-16. [PMID: 25310383 DOI: 10.1021/jm500862r] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Nonimmunosuppressive cyclophilin inhibitors have demonstrated efficacy for the treatment of hepatitis C infection (HCV). However, alisporivir, cyclosporin A, and most other cyclosporins are potent inhibitors of OATP1B1, MRP2, MDR1, and other important drug transporters. Reduction of the side chain hydrophobicity of the P4 residue preserves cyclophilin binding and antiviral potency while decreasing transporter inhibition. Representative inhibitor 33 (NIM258) is a less potent transporter inhibitor relative to previously described cyclosporins, retains anti-HCV activity in cell culture, and has an acceptable pharmacokinetic profile in rats and dogs. An X-ray structure of 33 bound to rat cyclophilin D is reported.
Collapse
Affiliation(s)
- Jiping Fu
- Novartis Institutes for Biomedical Research , 4560 Horton Street, Emeryville, California 94608, United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|