1
|
Liu LJ, Francisco KR, Sun YU, Serafim MSM, Amarasinghe DK, Teixeira TR, Lucero B, Kronenberger T, Elsayed W, Elwakeel H, Al-Hindy M, Almaliti J, Gerwick WH, O'Donoghue AJ, Caffrey CR. Carmaphycin B-Based Proteasome Inhibitors to Treat Human African Trypanosomiasis: Structure-Activity Relationship and In Vivo Efficacy. ACS Infect Dis 2024; 10:4182-4193. [PMID: 39589805 DOI: 10.1021/acsinfecdis.4c00441] [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] [Indexed: 11/28/2024]
Abstract
The proteasome is essential for eukaryotic cell proteostasis, and inhibitors of the 20S proteasome are progressing preclinically and clinically as antiparasitics. We screenedTrypanosoma brucei, the causative agent of human and animal African trypanosomiasis, in vitro with a set of 27 carmaphycin B analogs, irreversible epoxyketone inhibitors that were originally developed to inhibit thePlasmodium falciparum20S (Pf20S). The structure-activity relationship was distinct from that of the human c20S antitarget by the acceptance of d-amino acids at the P3 position of the peptidyl backbone to yield compounds with greatly decreased toxicity to human cells. For the three most selective compounds, binding to the Tb20S β5 catalytic subunit was confirmed by competition with a fluorescent activity-based probe. For one compound, J-80, with its P3 d-configuration, the differential binding to the parasite's β5 subunit was supported by both covalent and noncovalent docking analysis. Further, J-80 was equipotent against both Trypanosoma brucei gambiense and Trypanosoma brucei rhodesiense in vitro. In a mouse model of Stage 1 T. brucei infection, a single intraperitoneal (i.p.) dose of 40 mg/kg J-80 halted the growth of the parasite, and when given at 50 mg/kg i.p. twice daily for 5 days, parasitemia was decreased to below the detectable limit, with parasite recrudescence 48 h after the last dose. The in vivo proof of principle demonstrated by a potent, selective, and irreversible inhibitor of Tb20S reveals an alternative path to the development of kinetoplastid proteasome inhibitors that differs from the current focus on allosteric reversible inhibitors.
Collapse
Affiliation(s)
- Lawrence J Liu
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093, United States
| | - Karol R Francisco
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Yujie Uli Sun
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Mateus Sá Magalhães Serafim
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Microbiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG Brazil, 31270901
| | - Dilini K Amarasinghe
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Thaiz R Teixeira
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Bobby Lucero
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, California 92093, United States
| | - Thales Kronenberger
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry and Tübingen Center for Academic Drug Discovery (TüCAD2), Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio 70211, Finland
- Partner-site Tübingen, German Center for Infection Research (DZIF), Elfriede-Aulhorn-Str. 6, 72076 Tübingen, Germany
| | - Waad Elsayed
- Department of Medical Parasitology, Faculty of Medicine, Ain Shams University, Abassia, Cairo 1181, Egypt
| | - Hala Elwakeel
- Department of Medical Parasitology, Faculty of Medicine, Ain Shams University, Abassia, Cairo 1181, Egypt
| | - Momen Al-Hindy
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Jehad Almaliti
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Jordan, Amman 11942, Jordan
| | - William H Gerwick
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Anthony J O'Donoghue
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Conor R Caffrey
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
| |
Collapse
|
2
|
Malek N, Gladysz R, Stelmach N, Drag M. Targeting Microglial Immunoproteasome: A Novel Approach in Neuroinflammatory-Related Disorders. ACS Chem Neurosci 2024; 15:2532-2544. [PMID: 38970802 PMCID: PMC11258690 DOI: 10.1021/acschemneuro.4c00099] [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: 02/15/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/08/2024] Open
Abstract
It is widely acknowledged that the aging process is linked to the accumulation of damaged and misfolded proteins. This phenomenon is accompanied by a decrease in proteasome (c20S) activity, concomitant with an increase in immunoproteasome (i20S) activity. These changes can be attributed, in part, to the chronic neuroinflammation that occurs in brain tissues. Neuroinflammation is a complex process characterized by the activation of immune cells in the central nervous system (CNS) in response to injury, infection, and other pathological stimuli. In certain cases, this immune response becomes chronic, contributing to the pathogenesis of various neurological disorders, including chronic pain, Alzheimer's disease, Parkinson's disease, brain traumatic injury, and others. Microglia, the resident immune cells in the brain, play a crucial role in the neuroinflammatory response. Recent research has highlighted the involvement of i20S in promoting neuroinflammation, increased activity of which may lead to the presentation of self-antigens, triggering an autoimmune response against the CNS, exacerbating inflammation, and contributing to neurodegeneration. Furthermore, since i20S plays a role in breaking down accumulated proteins during inflammation within the cell body, any disruption in its activity could lead to a prolonged state of inflammation and subsequent cell death. Given the pivotal role of i20S in neuroinflammation, targeting this proteasome subtype has emerged as a potential therapeutic approach for managing neuroinflammatory diseases. This review delves into the mechanisms of neuroinflammation and microglia activation, exploring the potential of i20S inhibitors as a promising therapeutic strategy for managing neuroinflammatory disorders.
Collapse
Affiliation(s)
- Natalia Malek
- Department
of Chemical Biology and Bioimaging, Wroclaw
University of Science and Technology, ul. Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Radoslaw Gladysz
- Department
of Chemical Biology and Bioimaging, Wroclaw
University of Science and Technology, ul. Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Natalia Stelmach
- Department
of Chemical Biology and Bioimaging, Wroclaw
University of Science and Technology, ul. Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Marcin Drag
- Department
of Chemical Biology and Bioimaging, Wroclaw
University of Science and Technology, ul. Wybrzeze Wyspianskiego 27, 50-370 Wroclaw, Poland
| |
Collapse
|
3
|
Götz MG, Godwin K, Price R, Dorn R, Merrill-Steskal G, Klemmer W, Hansen H, Produturi G, Rocha M, Palmer M, Molacek L, Strater Z, Groll M. Macrocyclic Oxindole Peptide Epoxyketones-A Comparative Study of Macrocyclic Inhibitors of the 20S Proteasome. ACS Med Chem Lett 2024; 15:533-539. [PMID: 38628795 PMCID: PMC11017298 DOI: 10.1021/acsmedchemlett.4c00017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/22/2024] [Accepted: 03/22/2024] [Indexed: 04/19/2024] Open
Abstract
Peptide macrocycles have recently gained attention as protease inhibitors due to their metabolic stability and specificity. However, the development of peptide macrocycles with improved binding potency has so far been challenging. Here we present macrocyclic peptides derived from the clinically applied proteasome inhibitor carfilzomib with an oxindole group that mimics the natural product TMC-95A. Fluorescence kinetic activity assays reveal a high potency of the oxindole group (IC50 = 0.19 μM) compared with agents lacking this motif. X-ray structures of the ligands with the β5-subunit of the yeast 20S proteasome illustrate that the installed macrocycle forces strong hydrogen bonding of the oxindole group with β5-Gly23NH. Thus, the binding of our designed oxindole epoxyketones is entropically and enthalpically favored in contrast to more flexible proteasome inhibitors such as carfilzomib.
Collapse
Affiliation(s)
- Marion G. Götz
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| | - Kacey Godwin
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| | - Rachel Price
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| | - Robert Dorn
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| | | | - William Klemmer
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| | - Hunter Hansen
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| | - Gautam Produturi
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| | - Megan Rocha
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| | - Mathias Palmer
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| | - Lea Molacek
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| | - Zack Strater
- Department
of Chemistry, Whitman College, Walla Walla, Washington 99362, United States
| | - Michael Groll
- Technical
University of Munich, TUM School of Natural
Sciences, Department of Bioscience, Center for Protein Assemblies
(CPA), Ernst-Otto-Fischer
Strasse 8, 85748 Garching, Germany
| |
Collapse
|
4
|
Li S, He RC, Wu SG, Song Y, Zhang KL, Tang ML, Bei YR, Zhang T, Lu JB, Ma X, Jiang M, Qin LJ, Xu Y, Dong XH, Wu J, Dai X, Hu YW. LncRNA PSMB8-AS1 Instigates Vascular Inflammation to Aggravate Atherosclerosis. Circ Res 2024; 134:60-80. [PMID: 38084631 DOI: 10.1161/circresaha.122.322360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 11/20/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Increasing evidence suggests that long noncoding RNAs play significant roles in vascular biology and disease development. One such long noncoding RNA, PSMB8-AS1, has been implicated in the development of tumors. Nevertheless, the precise role of PSMB8-AS1 in cardiovascular diseases, particularly atherosclerosis, has not been thoroughly elucidated. Thus, the primary aim of this investigation is to assess the influence of PSMB8-AS1 on vascular inflammation and the initiation of atherosclerosis. METHODS We generated PSMB8-AS1 knockin and Apoe (Apolipoprotein E) knockout mice (Apoe-/-PSMB8-AS1KI) and global Apoe and proteasome subunit-β type-9 (Psmb9) double knockout mice (Apoe-/-Psmb9-/-). To explore the roles of PSMB8-AS1 and Psmb9 in atherosclerosis, we fed the mice with a Western diet for 12 weeks. RESULTS Long noncoding RNA PSMB8-AS1 is significantly elevated in human atherosclerotic plaques. Strikingly, Apoe-/-PSMB8-AS1KI mice exhibited increased atherosclerosis development, plaque vulnerability, and vascular inflammation compared with Apoe-/- mice. Moreover, the levels of VCAM1 (vascular adhesion molecule 1) and ICAM1 (intracellular adhesion molecule 1) were significantly upregulated in atherosclerotic lesions and serum of Apoe-/-PSMB8-AS1KI mice. Consistently, in vitro gain- and loss-of-function studies demonstrated that PSMB8-AS1 induced monocyte/macrophage adhesion to endothelial cells and increased VCAM1 and ICAM1 levels in a PSMB9-dependent manner. Mechanistic studies revealed that PSMB8-AS1 induced PSMB9 transcription by recruiting the transcription factor NONO (non-POU domain-containing octamer-binding protein) and binding to the PSMB9 promoter. PSMB9 (proteasome subunit-β type-9) elevated VCAM1 and ICAM1 expression via the upregulation of ZEB1 (zinc finger E-box-binding homeobox 1). Psmb9 deficiency decreased atherosclerotic lesion size, plaque vulnerability, and vascular inflammation in Apoe-/- mice in vivo. Importantly, endothelial overexpression of PSMB8-AS1-increased atherosclerosis and vascular inflammation were attenuated by Psmb9 knockout. CONCLUSIONS PSMB8-AS1 promotes vascular inflammation and atherosclerosis via the NONO/PSMB9/ZEB1 axis. Our findings support the development of new long noncoding RNA-based strategies to counteract atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Shu Li
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Run-Chao He
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Shao-Guo Wu
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, Guangdong, China (S.-G.W.)
| | - Yu Song
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Ke-Lan Zhang
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Mao-Lin Tang
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Yan-Rou Bei
- Laboratory Medicine Center (Y.-R.B.), Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ting Zhang
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Jin-Bo Lu
- Department of Peripheral Vascular Surgery, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen (J.-B.L.)
| | - Xin Ma
- Department of Anesthesiology (X.M.), Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Min Jiang
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Liang-Jun Qin
- Department of Pathology, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (L.J.Q.)
| | - Yudan Xu
- Laboratory Medicine Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China (Y.X.)
| | - Xian-Hui Dong
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Jia Wu
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
| | - Xiaoyan Dai
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangdong, China (X.D.)
- Clinical Research Institute, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, China (X.D.)
| | - Yan-Wei Hu
- Department of Clinical Laboratory, Guangzhou Women & Children Medical Center, Guangzhou Medical University, Guangdong, China (S.L., R.-C.H., Y.S., K.-L.Z., M.-L.T., T.Z., M.J., X.-H.D., J.W., Y.-W.H.)
- Department of Laboratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China (Y.-W.H.)
| |
Collapse
|
5
|
Culletta G, Tutone M, Ettari R, Perricone U, Di Chio C, Almerico AM, Zappalà M. Virtual Screening Strategy and In Vitro Tests to Identify New Inhibitors of the Immunoproteasome. Int J Mol Sci 2023; 24:10504. [PMID: 37445688 DOI: 10.3390/ijms241310504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Immunoproteasome inhibition is a promising strategy for the treatment of hematological malignancies, autoimmune diseases, and inflammatory diseases. The design of non-covalent inhibitors of the immunoproteasome β1i/β5i catalytic subunits could be a novel approach to avoid the drawbacks of the known covalent inhibitors, such as toxicity due to off-target binding. In this work, we report the biological evaluation of thirty-four compounds selected from a commercially available collection. These hit compounds are the outcomes of a virtual screening strategy including a dynamic pharmacophore modeling approach onto the β1i subunit and a pharmacophore/docking approach onto the β5i subunit. The computational studies were first followed by in vitro enzymatic assays at 100 μM. Only compounds capable of inhibiting the enzymatic activity by more than 50% were characterized in detail using Tian continuous assays, determining the dissociation constant (Ki) of the non-covalent complex where Ki is also the measure of the binding affinity. Seven out of thirty-four hits showed to inhibit β1i and/or β5i subunit. Compound 3 is the most active on the β1i subunit with Ki = 11.84 ± 1.63 µM, and compound 17 showed Ki = 12.50 ± 0.77 µM on the β5i subunit. Compound 2 showed inhibitory activity on both subunits (Ki = 12.53 ± 0.18 and Ki = 31.95 ± 0.81 on the β1i subunit and β5i subunit, respectively). The induced fit docking analysis revealed interactions with Thr1 and Phe31 of β1i subunit and that represent new key residues as reported in our previous work. Onto β5i subunit, it interacts with the key residues Thr1, Thr21, and Tyr169. This last hit compound identified represents an interesting starting point for further optimization of β1i/β5i dual inhibitors of the immunoproteasome.
Collapse
Affiliation(s)
- Giulia Culletta
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università di Messina, Viale Annunziata, 98168 Messina, Italy
| | - Marco Tutone
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Roberta Ettari
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università di Messina, Viale Annunziata, 98168 Messina, Italy
| | - Ugo Perricone
- Drug Discovery Unit, Fondazione Ri.MED, 90133 Palermo, Italy
| | - Carla Di Chio
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università di Messina, Viale Annunziata, 98168 Messina, Italy
| | - Anna Maria Almerico
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Maria Zappalà
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, Università di Messina, Viale Annunziata, 98168 Messina, Italy
| |
Collapse
|
6
|
Almaliti J, Fajtová P, Calla J, LaMonte GM, Feng M, Rocamora F, Ottilie S, Glukhov E, Boura E, Suhandynata RT, Momper JD, Gilson MK, Winzeler EA, Gerwick WH, O'Donoghue AJ. Development of Potent and Highly Selective Epoxyketone-Based Plasmodium Proteasome Inhibitors. Chemistry 2023; 29:e202203958. [PMID: 36617500 PMCID: PMC10894724 DOI: 10.1002/chem.202203958] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/03/2023] [Accepted: 01/03/2023] [Indexed: 01/10/2023]
Abstract
Here, we present remarkable epoxyketone-based proteasome inhibitors with low nanomolar in vitro potency for blood-stage Plasmodium falciparum and low cytotoxicity for human cells. Our best compound has more than 2,000-fold greater selectivity for erythrocytic-stage P. falciparum over HepG2 and H460 cells, which is largely driven by the accommodation of the parasite proteasome for a D-amino acid in the P3 position and the preference for a difluorobenzyl group in the P1 position. We isolated the proteasome from P. falciparum cell extracts and determined that the best compound is 171-fold more potent at inhibiting the β5 subunit of P. falciparum proteasome when compared to the same subunit of the human constitutive proteasome. These compounds also significantly reduce parasitemia in a P. berghei mouse infection model and prolong survival of animals by an average of 6 days. The current epoxyketone inhibitors are ideal starting compounds for orally bioavailable anti-malarial drugs.
Collapse
Affiliation(s)
- Jehad Almaliti
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
- Department Pharmaceutical Sciences, College of Pharmacy, University of Jordan, Amman, 11942, Jordan
| | - Pavla Fajtová
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 16610, Prague, Czech Republic
| | - Jaeson Calla
- Department of Pediatrics, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Gregory M LaMonte
- Department of Pediatrics, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Mudong Feng
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Frances Rocamora
- Department of Pediatrics, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Sabine Ottilie
- Department of Pediatrics, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
- Calibr, a division of The Scripps Research Institute, 11119 N Torrey Pines Rd, La Jolla, California, 92093, USA
| | - Evgenia Glukhov
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Evzen Boura
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, 16610, Prague, Czech Republic
| | - Raymond T Suhandynata
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Jeremiah D Momper
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Michael K Gilson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Elizabeth A Winzeler
- Department of Pediatrics, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - William H Gerwick
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
- Center for Marine Biotechnology and Biomedicine, Scripps Institution of Oceanography, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| | - Anthony J O'Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, California, 92093, USA
| |
Collapse
|
7
|
Allardyce D, Adu Mantey P, Szalecka M, Nkwo R, Loizidou EZ. Identification of a new class of proteasome inhibitors based on a naphthyl-azotricyclic-urea-phenyl scaffold. RSC Med Chem 2023; 14:573-582. [PMID: 36970145 PMCID: PMC10034219 DOI: 10.1039/d2md00404f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 02/06/2023] [Indexed: 02/09/2023] Open
Abstract
Proteasomes play an important role in protein degradation and regulation of many cellular pathways by maintaining protein balance. Inhibitors of proteasomes disrupt this balance affecting proteins that are key in malignancies and as such have found applications in the treatment of multiple myeloma and mantle cell lymphoma. However, resistance mechanisms have been reported for these proteasome inhibitors including mutations at the β5 site which necessitates the constant development of new inhibitors. In this work, we report the identification of a new class of proteasome inhibitors, polycyclic molecules bearing a naphthyl-azotricyclic-urea-phenyl scaffold, from screening of the ZINC library of natural products. The most potent of these compounds showed evidence of dose dependency through proteasome assays with IC50 values in the low micromolar range, and kinetic analysis revealed competitive binding at the β5c site with an estimated inhibition constant, K i, of 1.15 μM. Inhibition was also shown for the β5i site of the immunoproteasome at levels similar to those of the constitutive proteasome. Structure-activity relationship studies identified the naphthyl substituent to be crucial for activity and this was attributed to enhanced hydrophobic interactions within β5c. Further to this, halogen substitution within the naphthyl ring enhanced the activity and allowed for π-π interactions with Y169 in β5c and Y130 and F124 in β5i. The combined data highlight the importance of hydrophobic and halogen interactions in β5 binding and assist in the design of next generation inhibitors of proteasomes.
Collapse
Affiliation(s)
- Duncan Allardyce
- Faculty of Science and Technology, Department of Natural Sciences, Middlesex University The Burroughs London NW4 4BT UK
| | - Priscilla Adu Mantey
- Faculty of Science and Technology, Department of Natural Sciences, Middlesex University The Burroughs London NW4 4BT UK
| | - Monika Szalecka
- Faculty of Science and Technology, Department of Natural Sciences, Middlesex University The Burroughs London NW4 4BT UK
| | - Robert Nkwo
- Faculty of Science and Technology, Department of Natural Sciences, Middlesex University The Burroughs London NW4 4BT UK
| | - Eriketi Z Loizidou
- Faculty of Science and Technology, Department of Natural Sciences, Middlesex University The Burroughs London NW4 4BT UK
| |
Collapse
|
8
|
Wang X, Zhang H, Wang Y, Bramasole L, Guo K, Mourtada F, Meul T, Hu Q, Viteri V, Kammerl I, Konigshoff M, Lehmann M, Magg T, Hauck F, Fernandez IE, Meiners S. DNA sensing via the cGAS/STING pathway activates the immunoproteasome and adaptive T-cell immunity. EMBO J 2023; 42:e110597. [PMID: 36912165 PMCID: PMC10106989 DOI: 10.15252/embj.2022110597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/09/2023] [Accepted: 02/20/2023] [Indexed: 03/14/2023] Open
Abstract
The immunoproteasome is a specialized type of proteasome involved in MHC class I antigen presentation, antiviral adaptive immunity, autoimmunity, and is also part of a broader response to stress. Whether the immunoproteasome is regulated by DNA stress, however, is not known. We here demonstrate that mitochondrial DNA stress upregulates the immunoproteasome and MHC class I antigen presentation pathway via cGAS/STING/type I interferon signaling resulting in cell autonomous activation of CD8+ T cells. The cGAS/STING-induced adaptive immune response is also observed in response to genomic DNA and is conserved in epithelial and mesenchymal cells of mice and men. In patients with idiopathic pulmonary fibrosis, chronic activation of the cGAS/STING-induced adaptive immune response in aberrant lung epithelial cells concurs with CD8+ T-cell activation in diseased lungs. Genetic depletion of the immunoproteasome and specific immunoproteasome inhibitors counteract DNA stress induced cytotoxic CD8+ T-cell activation. Our data thus unravel cytoplasmic DNA sensing via the cGAS/STING pathway as an activator of the immunoproteasome and CD8+ T cells. This represents a novel potential pathomechanism for pulmonary fibrosis that opens new therapeutic perspectives.
Collapse
Affiliation(s)
- Xinyuan Wang
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany.,State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Huabin Zhang
- Neurosurgical Research, Department of Neurosurgery, University Hospital and Walter-Brendel-Centre of Experimental Medicine, Faculty of Medicine, Ludwig-Maximilians-University, Munich, Germany.,The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yuqin Wang
- Research Center Borstel/Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Laylan Bramasole
- Research Center Borstel/Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Kai Guo
- Research Center Borstel/Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Fatima Mourtada
- Research Center Borstel/Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Thomas Meul
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany
| | - Qianjiang Hu
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), University Hospital Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Valeria Viteri
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany
| | - Ilona Kammerl
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany
| | - Melanie Konigshoff
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), University Hospital Grosshadern, Ludwig-Maximilians-University, Munich, Germany.,Division of Pulmonary, Allergy and Critical Care Medicine, School of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mareike Lehmann
- Research Unit Lung Repair and Regeneration, Helmholtz Zentrum München, German Research Center for Environmental Health, Member of the German Center of Lung Research (DZL), University Hospital Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Thomas Magg
- Division of Pediatric Immunology and Rheumatology, Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Fabian Hauck
- Division of Pediatric Immunology and Rheumatology, Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Isis E Fernandez
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany.,Department of Medicine V, University Hospital, LMU Munich, Munich, Germany
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), Member of the German Center for Lung Research (DZL), University Hospital, Ludwig-Maximilians University, Helmholtz Zentrum München, Munich, Germany.,Research Center Borstel/Leibniz Lung Center, Borstel, Germany.,Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|
9
|
Targeting immunoproteasome in neurodegeneration: A glance to the future. Pharmacol Ther 2023; 241:108329. [PMID: 36526014 DOI: 10.1016/j.pharmthera.2022.108329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/02/2022] [Accepted: 12/05/2022] [Indexed: 12/15/2022]
Abstract
The immunoproteasome is a specialized form of proteasome equipped with modified catalytic subunits that was initially discovered to play a pivotal role in MHC class I antigen processing and immune system modulation. However, over the last years, this proteolytic complex has been uncovered to serve additional functions unrelated to antigen presentation. Accordingly, it has been proposed that immunoproteasome synergizes with canonical proteasome in different cell types of the nervous system, regulating neurotransmission, metabolic pathways and adaptation of the cells to redox or inflammatory insults. Hence, studying the alterations of immunoproteasome expression and activity is gaining research interest to define the dynamics of neuroinflammation as well as the early and late molecular events that are likely involved in the pathogenesis of a variety of neurological disorders. Furthermore, these novel functions foster the perspective of immunoproteasome as a potential therapeutic target for neurodegeneration. In this review, we provide a brain and retina-wide overview, trying to correlate present knowledge on structure-function relationships of immunoproteasome with the variety of observed neuro-modulatory functions.
Collapse
|
10
|
The dichotomous role of immunoproteasome in cancer: Friend or foe? Acta Pharm Sin B 2022; 13:1976-1989. [DOI: 10.1016/j.apsb.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/21/2022] [Accepted: 10/07/2022] [Indexed: 11/08/2022] Open
|
11
|
Immunoproteasome Activity in Chronic Lymphocytic Leukemia as a Target of the Immunoproteasome-Selective Inhibitors. Cells 2022; 11:cells11050838. [PMID: 35269460 PMCID: PMC8909520 DOI: 10.3390/cells11050838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 12/21/2022] Open
Abstract
Targeting proteasome with proteasome inhibitors (PIs) is an approved treatment strategy in multiple myeloma that has also been explored pre-clinically and clinically in other hematological malignancies. The approved PIs target both the constitutive and the immunoproteasome, the latter being present predominantly in cells of lymphoid origin. Therapeutic targeting of the immunoproteasome in cells with sole immunoproteasome activity may be selectively cytotoxic in malignant cells, while sparing the non-lymphoid tissues from the on-target PIs toxicity. Using activity-based probes to assess the proteasome activity profile and correlating it with the cytotoxicity assays, we identified B-cell chronic lymphocytic leukemia (B-CLL) to express predominantly immunoproteasome activity, which is associated with high sensitivity to approved proteasome inhibitors and, more importantly, to the immunoproteasome selective inhibitors LU005i and LU035i, targeting all immunoproteasome active subunits or only the immunoproteasome β5i, respectively. At the same time, LU102, a proteasome β2 inhibitor, sensitized B-CLL or immunoproteasome inhibitor-inherently resistant primary cells of acute myeloid leukemia, B-cell acute lymphoblastic leukemia, multiple myeloma and plasma cell leukemia to low doses of LU035i. The immunoproteasome thus represents a novel therapeutic target, which warrants further testing with clinical stage immunoproteasome inhibitors in monotherapy or in combinations.
Collapse
|
12
|
Kisselev AF. Site-Specific Proteasome Inhibitors. Biomolecules 2021; 12:54. [PMID: 35053202 PMCID: PMC8773591 DOI: 10.3390/biom12010054] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 12/14/2022] Open
Abstract
Proteasome is a multi-subunit protein degradation machine, which plays a key role in the maintenance of protein homeostasis and, through degradation of regulatory proteins, in the regulation of numerous cell functions. Proteasome inhibitors are essential tools for biomedical research. Three proteasome inhibitors, bortezomib, carfilzomib, and ixazomib are approved by the FDA for the treatment of multiple myeloma; another inhibitor, marizomib, is undergoing clinical trials. The proteolytic core of the proteasome has three pairs of active sites, β5, β2, and β1. All clinical inhibitors and inhibitors that are widely used as research tools (e.g., epoxomicin, MG-132) inhibit multiple active sites and have been extensively reviewed in the past. In the past decade, highly specific inhibitors of individual active sites and the distinct active sites of the lymphoid tissue-specific immunoproteasome have been developed. Here, we provide a comprehensive review of these site-specific inhibitors of mammalian proteasomes and describe their utilization in the studies of the biology of the active sites and their roles as drug targets for the treatment of different diseases.
Collapse
Affiliation(s)
- Alexei F Kisselev
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL 36849, USA
| |
Collapse
|
13
|
A Cell-Based Platform for the Investigation of Immunoproteasome Subunit β5i Expression and Biology of β5i-Containing Proteasomes. Cells 2021; 10:cells10113049. [PMID: 34831272 PMCID: PMC8616536 DOI: 10.3390/cells10113049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 10/24/2021] [Accepted: 11/02/2021] [Indexed: 11/17/2022] Open
Abstract
The degradation of most intracellular proteins is a dynamic and tightly regulated process performed by proteasomes. To date, different forms of proteasomes have been identified. Currently the role of non-constitutive proteasomes (immunoproteasomes (iPs) and intermediate proteasomes (intPs)) has attracted special attention. Here, using a CRISPR-Cas9 nickase technology, four cell lines: histiocytic lymphoma, colorectal adenocarcinoma, cervix adenocarcinoma, and hepatocarcinoma were modified to express proteasomes with mCherry-tagged β5i subunit, which is a catalytic subunit of iPs and intPs. Importantly, the expression of the chimeric gene in modified cells is under the control of endogenous regulatory mechanisms and is increased following IFN-γ and/or TNF-α stimulation. Fluorescent proteasomes retain catalytic activity and are distributed within the nucleus and cytoplasm. RNAseq reveals marginal differences in gene expression profiles between the modified and wild-type cell lines. Predominant metabolic pathways and patterns of expressed receptors were identified for each cell line. Using established cell lines, we demonstrated that anti-cancer drugs Ruxolitinib, Vincristine and Gefitinib stimulated the expression of β5i-containing proteasomes, which might affect disease prognosis. Taken together, obtained cell lines can be used as a platform for real-time studies of immunoproteasome gene expression, localization of iPs and intPs, interaction of non-constitutive proteasomes with other proteins, proteasome trafficking and many other aspects of proteasome biology in living cells. Moreover, the established platform might be especially useful for fast and large-scale experiments intended to evaluate the effects of different conditions including treatment with various drugs and compounds on the proteasome pool.
Collapse
|
14
|
High Immunoproteasome Activity and sXBP1 in Pediatric Precursor B-ALL Predicts Sensitivity towards Proteasome Inhibitors. Cells 2021; 10:cells10112853. [PMID: 34831075 PMCID: PMC8616377 DOI: 10.3390/cells10112853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/30/2022] Open
Abstract
Proteasome inhibitors (PIs) are approved backbone treatments in multiple myeloma. More recently, inhibition of proteasome activity with the PI bortezomib has been clinically evaluated as a novel treatment strategy in pediatric acute lymphoblastic leukemia (ALL). However, we lack a marker that could identify ALL patients responding to PI-based therapy. By using a set of activity-based proteasome probes in conjunction with cytotoxicity assays, we show that B-cell precursor ALL (BCP-ALL), in contrast to T-ALL, demonstrates an increased activity of immunoproteasome over constitutive proteasome, which correlates with high ex vivo sensitivity to the PIs bortezomib and ixazomib. The novel selective PI LU015i-targeting immunoproteasome β5i induces cytotoxicity in BCP-ALL containing high β5i activity, confirming immunoproteasome activity as a novel therapeutic target in BCP-ALL. At the same time, cotreatment with β2-selective proteasome inhibitors can sensitize T-ALL to currently available PIs, as well as to β5i selective PI. In addition, levels of total and spliced forms of XBP1 differ between BCP-ALL and T-ALL, and only in BCP-ALL does high-spliced XBP1 correlate with sensitivity to bortezomib. Thus, in BCP-ALL, high immunoproteasome activity may serve as a predictive marker for PI-based treatment options, potentially combined with XBP1 analyses.
Collapse
|
15
|
Kammerl IE, Hardy S, Flexeder C, Urmann A, Peierl J, Wang Y, Vosyka O, Frankenberger M, Milger K, Behr J, Koch A, Merl-Pham J, Hauck SM, Pilette C, Schulz H, Meiners S. Activation of immune cell proteasomes in peripheral blood of smokers and COPD patients - implications for therapy. Eur Respir J 2021; 59:13993003.01798-2021. [PMID: 34561290 PMCID: PMC8891681 DOI: 10.1183/13993003.01798-2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/19/2021] [Indexed: 11/05/2022]
Abstract
Immune cells contain a specialised type of proteasome, i.e. the immunoproteasome, which is required for intracellular protein degradation. Immunoproteasomes are key regulators of immune cell differentiation, inflammatory activation and autoimmunity. Immunoproteasome function in peripheral immune cells might be altered by smoking and in COPD thereby affecting immune cell responses.We here analysed the expression and activity of proteasome complexes in peripheral blood mononuclear cells (PBMC) isolated from healthy male young smokers as well as from patients with severe COPD and compared them to matching controls. Proteasome expression was upregulated in COPD patients as assessed by RT-qPCR and mass spectrometry-based proteomics analysis. Proteasome activity was quantified using activity-based probes and native gel analysis. We observed distinct activation of immunoproteasomes in the peripheral blood cells of young male smokers and severely ill COPD patients. Native gel analysis and linear regression modeling confirmed robust activation and elevated assembly of 20S proteasomes, which correlated significantly with reduced lung function parameters in COPD patients. The immunoproteasome was distinctly activated in COPD patients upon inflammatory cytokine stimulation of PBMCs in vitro Inhibition of the immunoproteasome reduced proinflammatory cytokine expression in COPD-derived blood immune cells.Given the crucial role of chronic inflammatory signalling and the emerging involvement of autoimmune responses in COPD, therapeutic targeting of the immunoproteasome might represent a novel therapeutic concept for COPD.
Collapse
Affiliation(s)
- Ilona E Kammerl
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Sophie Hardy
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany.,Cliniques universitaires Saint-Luc, department of pulmonology, and Institute of Experimental and Clinical Research (IREC), Pole of pulmonology, ENT and dermatology, Université catholique de Louvain, Brussels, Belgium
| | - Claudia Flexeder
- Institute of Epidemiology, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany
| | - Andrea Urmann
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Julia Peierl
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Yuqin Wang
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Oliver Vosyka
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Marion Frankenberger
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany.,Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Katrin Milger
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany.,Department of Medicine V, University Hospital, LMU, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Jürgen Behr
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany.,Department of Medicine V, University Hospital, LMU, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Andrea Koch
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany.,Dept. of Pneumology, Teaching Hospital Pyhrn-Eisenwurzen Klinikum Steyr, Austria
| | - Juliane Merl-Pham
- Research Unit Protein Science, Metabolomics and Proteomics Core, Helmholtz Zentrum München, Munich, Germany
| | - Stefanie M Hauck
- Research Unit Protein Science, Metabolomics and Proteomics Core, Helmholtz Zentrum München, Munich, Germany
| | - Charles Pilette
- Cliniques universitaires Saint-Luc, department of pulmonology, and Institute of Experimental and Clinical Research (IREC), Pole of pulmonology, ENT and dermatology, Université catholique de Louvain, Brussels, Belgium
| | - Holger Schulz
- Institute of Epidemiology, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Neuherberg, Germany
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig-Maximilians- University, Helmholtz Zentrum München, Member of the German Center for Lung Research (DZL), Munich, Germany
| |
Collapse
|
16
|
Pseudopeptides with aldehyde or vinylsulfone warheads: Synthesis and antiproteasomal activity. Bioorg Chem 2021; 115:105228. [PMID: 34371374 DOI: 10.1016/j.bioorg.2021.105228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/26/2021] [Accepted: 07/28/2021] [Indexed: 11/22/2022]
Abstract
The comparative study of new proteasome inhibitors based on salicylic acid-modified pseudo-tripeptides terminated with aldehyde or vinylsulfone is presented. We described the synthesis of 11 pairs of pseudopeptides and their properties related to the proteasome inhibition were determined. The effects of integrated amino acids (combinations of leucine, phenylalanine, tryptophan, proline, cyclohexylalanine or norleucine residues) on the activity of the proteasome were investigated. Compounds preferentially inhibited the chymotrypsin β5-subunit of the proteasome in cell-based assays compared with the β1- and β2-subunits, with IC50 values in mid-nanomolar ranges being obtained for the most active members. Our comparative study demonstrated that aldehydes were able to inhibit the proteasome in cells more effectively than vinylsulfones. These results were corroborated by the accumulation of polyubiquitinated proteins in treated cells, GFP accumulation in a reporter cell line and the ability of new compounds to induce apoptotic cell death.
Collapse
|
17
|
A Nut for Every Bolt: Subunit-Selective Inhibitors of the Immunoproteasome and Their Therapeutic Potential. Cells 2021; 10:cells10081929. [PMID: 34440698 PMCID: PMC8394499 DOI: 10.3390/cells10081929] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 12/15/2022] Open
Abstract
At the heart of the ubiquitin-proteasome system, the 20S proteasome core particle (CP) breaks down the majority of intracellular proteins tagged for destruction. Thereby, the CP controls many cellular processes including cell cycle progression and cell signalling. Inhibitors of the CP can suppress these essential biological pathways, resulting in cytotoxicity, an effect that is beneficial for the treatment of certain blood cancer patients. During the last decade, several preclinical studies demonstrated that selective inhibition of the immunoproteasome (iCP), one of several CP variants in mammals, suppresses autoimmune diseases without inducing toxic side effects. These promising findings led to the identification of natural and synthetic iCP inhibitors with distinct chemical structures, varying potency and subunit selectivity. This review presents the most prominent iCP inhibitors with respect to possible scientific and medicinal applications, and discloses recent trends towards pan-immunoproteasome reactive inhibitors that cumulated in phase II clinical trials of the lead compound KZR-616 for chronic inflammations.
Collapse
|
18
|
Immunoproteasome Function in Normal and Malignant Hematopoiesis. Cells 2021; 10:cells10071577. [PMID: 34206607 PMCID: PMC8305381 DOI: 10.3390/cells10071577] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/16/2021] [Accepted: 06/16/2021] [Indexed: 12/19/2022] Open
Abstract
The ubiquitin-proteasome system (UPS) is a central part of protein homeostasis, degrading not only misfolded or oxidized proteins but also proteins with essential functions. The fact that a healthy hematopoietic system relies on the regulation of protein homeostasis and that alterations in the UPS can lead to malignant transformation makes the UPS an attractive therapeutic target for the treatment of hematologic malignancies. Herein, inhibitors of the proteasome, the last and most important component of the UPS enzymatic cascade, have been approved for the treatment of these malignancies. However, their use has been associated with side effects, drug resistance, and relapse. Inhibitors of the immunoproteasome, a proteasomal variant constitutively expressed in the cells of hematopoietic origin, could potentially overcome the encountered problems of non-selective proteasome inhibition. Immunoproteasome inhibitors have demonstrated their efficacy and safety against inflammatory and autoimmune diseases, even though their development for the treatment of hematologic malignancies is still in the early phases. Various immunoproteasome inhibitors have shown promising preliminary results in pre-clinical studies, and one inhibitor is currently being investigated in clinical trials for the treatment of multiple myeloma. Here, we will review data on immunoproteasome function and inhibition in hematopoietic cells and hematologic cancers.
Collapse
|
19
|
Jenkins TW, Downey-Kopyscinski SL, Fields JL, Rahme GJ, Colley WC, Israel MA, Maksimenko AV, Fiering SN, Kisselev AF. Activity of immunoproteasome inhibitor ONX-0914 in acute lymphoblastic leukemia expressing MLL-AF4 fusion protein. Sci Rep 2021; 11:10883. [PMID: 34035431 PMCID: PMC8149845 DOI: 10.1038/s41598-021-90451-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/12/2021] [Indexed: 11/08/2022] Open
Abstract
Proteasome inhibitors bortezomib and carfilzomib are approved for the treatment of multiple myeloma and mantle cell lymphoma and have demonstrated clinical efficacy for the treatment of acute lymphoblastic leukemia (ALL). The t(4;11)(q21;q23) chromosomal translocation that leads to the expression of MLL-AF4 fusion protein and confers a poor prognosis, is the major cause of infant ALL. This translocation sensitizes tumor cells to proteasome inhibitors, but toxicities of bortezomib and carfilzomib may limit their use in pediatric patients. Many of these toxicities are caused by on-target inhibition of proteasomes in non-lymphoid tissues (e.g., heart muscle, gut, testicles). We found that MLL-AF4 cells express high levels of lymphoid tissue-specific immunoproteasomes and are sensitive to pharmacologically relevant concentrations of specific immunoproteasome inhibitor ONX-0914, even in the presence of stromal cells. Inhibition of multiple active sites of the immunoproteasomes was required to achieve cytotoxicity against ALL. ONX-0914, an inhibitor of LMP7 (ß5i) and LMP2 (ß1i) sites of the immunoproteasome, and LU-102, inhibitor of proteasome ß2 sites, exhibited synergistic cytotoxicity. Treatment with ONX-0914 significantly delayed the growth of orthotopic ALL xenograft tumors in mice. T-cell ALL lines were also sensitive to pharmacologically relevant concentrations of ONX-0914. This study provides a strong rationale for testing clinical stage immunoproteasome inhibitors KZ-616 and M3258 in ALL.
Collapse
Affiliation(s)
- Tyler W Jenkins
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, PRB, 720 S. Donahue Dr., Auburn, AL, 36849, USA
| | - Sondra L Downey-Kopyscinski
- Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- SLDK - Rancho Biosciences, San Diego, CA, USA
- GJR- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- WCC - ScribeAmerica, Huntsville Hospital, Huntsville, AL, USA
- MAI- Israel Cancer Research Fund, New York, NY, USA
| | - Jennifer L Fields
- Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Gilbert J Rahme
- Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- SLDK - Rancho Biosciences, San Diego, CA, USA
- GJR- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- WCC - ScribeAmerica, Huntsville Hospital, Huntsville, AL, USA
- MAI- Israel Cancer Research Fund, New York, NY, USA
| | - William C Colley
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, PRB, 720 S. Donahue Dr., Auburn, AL, 36849, USA
- SLDK - Rancho Biosciences, San Diego, CA, USA
- GJR- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- WCC - ScribeAmerica, Huntsville Hospital, Huntsville, AL, USA
- MAI- Israel Cancer Research Fund, New York, NY, USA
| | - Mark A Israel
- Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- SLDK - Rancho Biosciences, San Diego, CA, USA
- GJR- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- WCC - ScribeAmerica, Huntsville Hospital, Huntsville, AL, USA
- MAI- Israel Cancer Research Fund, New York, NY, USA
| | - Andrey V Maksimenko
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, PRB, 720 S. Donahue Dr., Auburn, AL, 36849, USA
| | - Steven N Fiering
- Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
- Department of Microbiology and Immunology, Geisel School of Medicine, Dartmouth College, Lebanon, NH, USA
| | - Alexei F Kisselev
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, PRB, 720 S. Donahue Dr., Auburn, AL, 36849, USA.
| |
Collapse
|
20
|
Li X, Hong D, Zhang M, Xu L, Zhou Y, Li J, Liu T. Development of peptide epoxyketones as selective immunoproteasome inhibitors. Eur J Med Chem 2021; 221:113556. [PMID: 34087498 DOI: 10.1016/j.ejmech.2021.113556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 05/06/2021] [Accepted: 05/14/2021] [Indexed: 11/15/2022]
Abstract
A series of epoxyketone analogues with varying N-caps and P3-configurations were designed, synthesized and evaluated. We found that D-Ala in P3 was crucial for β5i selectivity over β5c. Notably, compounds 20j (β5i IC50 = 26.0 nM, 25-fold selectivity) and 20l (β5i IC50 = 25.1 nM, 24-fold selectivity) with the D-configuration at P3 were the most selective inhibitors. Although 20j and 20l showed only moderate anti-proliferative activity against RPMI-8226 and MM.1S cell lines, based on our experiments, it indicates that the inhibition of β5i alone is not sufficient to exert anticancer effects and may rely on the complementary inhibition of β1i, β5c and β5i. These data further increase our understanding of immunoproteasome inhibitors in hematologic malignancies.
Collapse
Affiliation(s)
- Xuemei Li
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China
| | - Duidui Hong
- Jiangsu Shengdia Industrial Co. Ltd., NO. 161 Shaoxing Road, Xiacheng District, Hangzhou, 310004, PR China
| | - Mengmeng Zhang
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China
| | - Lei Xu
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China; Zhongshan Institute of Drug Discovery, Institution for Drug Discovery Innovation, Chinese Academy of Science, Zhongshan, 528400, PR China
| | - Yubo Zhou
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China; Zhongshan Institute of Drug Discovery, Institution for Drug Discovery Innovation, Chinese Academy of Science, Zhongshan, 528400, PR China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, PR China.
| | - Jia Li
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, PR China; Zhongshan Institute of Drug Discovery, Institution for Drug Discovery Innovation, Chinese Academy of Science, Zhongshan, 528400, PR China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing, 100049, PR China.
| | - Tao Liu
- ZJU-ENS Joint Laboratory of Medicinal Chemistry, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, PR China.
| |
Collapse
|
21
|
Kim HR, Tagirasa R, Yoo E. Covalent Small Molecule Immunomodulators Targeting the Protease Active Site. J Med Chem 2021; 64:5291-5322. [PMID: 33904753 DOI: 10.1021/acs.jmedchem.1c00172] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cells of the immune system utilize multiple proteases to regulate cell functions and orchestrate innate and adaptive immune responses. Dysregulated protease activities are implicated in many immune-related disorders; thus, protease inhibitors have been actively investigated for pharmaceutical development. Although historically considered challenging with concerns about toxicity, compounds that covalently modify the protease active site represent an important class of agents, emerging not only as chemical probes but also as approved drugs. Here, we provide an overview of technologies useful for the study of proteases with the focus on recent advances in chemoproteomic methods and screening platforms. By highlighting covalent inhibitors that have been designed to target immunomodulatory proteases, we identify opportunities for the development of small molecule immunomodulators.
Collapse
Affiliation(s)
- Hong-Rae Kim
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Ravichandra Tagirasa
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Euna Yoo
- Chemical Biology Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| |
Collapse
|
22
|
Kollár L, Gobec M, Szilágyi B, Proj M, Knez D, Ábrányi-Balogh P, Petri L, Imre T, Bajusz D, Ferenczy GG, Gobec S, Keserű GM, Sosič I. Discovery of selective fragment-sized immunoproteasome inhibitors. Eur J Med Chem 2021; 219:113455. [PMID: 33894528 DOI: 10.1016/j.ejmech.2021.113455] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/14/2021] [Accepted: 04/05/2021] [Indexed: 11/26/2022]
Abstract
Proteasomes contribute to maintaining protein homeostasis and their inhibition is beneficial in certain types of cancer and in autoimmune diseases. However, the inhibition of the proteasomes in healthy cells leads to unwanted side-effects and significant effort has been made to identify inhibitors specific for the immunoproteasome, especially to treat diseases which manifest increased levels and activity of this proteasome isoform. Here, we report our efforts to discover fragment-sized inhibitors of the human immunoproteasome. The screening of an in-house library of structurally diverse fragments resulted in the identification of benzo[d]oxazole-2(3H)-thiones, benzo[d]thiazole-2(3H)-thiones, benzo[d]imidazole-2(3H)-thiones, and 1-methylbenzo[d]imidazole-2(3H)-thiones (with a general term benzoXazole-2(3H)-thiones) as inhibitors of the chymotrypsin-like (β5i) subunit of the immunoproteasome. A subsequent structure-activity relationship study provided us with an insight regarding growing vectors. Binding to the β5i subunit was shown and selectivity against the β5 subunit of the constitutive proteasome was determined. Thorough characterization of these compounds suggested that they inhibit the immunoproteasome by forming a disulfide bond with the Cys48 available specifically in the β5i active site. To obtain fragments with biologically more tractable covalent interactions, we performed a warhead scan, which yielded benzoXazole-2-carbonitriles as promising starting points for the development of selective immunoproteasome inhibitors with non-peptidic scaffolds.
Collapse
Affiliation(s)
- Levente Kollár
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117, Budapest, Hungary
| | - Martina Gobec
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, SI-1000, Ljubljana, Slovenia
| | - Bence Szilágyi
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117, Budapest, Hungary
| | - Matic Proj
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, SI-1000, Ljubljana, Slovenia
| | - Damijan Knez
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, SI-1000, Ljubljana, Slovenia
| | - Péter Ábrányi-Balogh
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117, Budapest, Hungary
| | - László Petri
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117, Budapest, Hungary
| | - Tímea Imre
- MS Metabolomics Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117, Budapest, Hungary
| | - Dávid Bajusz
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117, Budapest, Hungary
| | - György G Ferenczy
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117, Budapest, Hungary
| | - Stanislav Gobec
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, SI-1000, Ljubljana, Slovenia
| | - György M Keserű
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117, Budapest, Hungary.
| | - Izidor Sosič
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva cesta 7, SI-1000, Ljubljana, Slovenia.
| |
Collapse
|
23
|
Synthesis of macrocyclic α-ketoamide as a selective and reversible immunoproteasome inhibitor. Med Chem Res 2021. [DOI: 10.1007/s00044-020-02678-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
24
|
Maltsev A, Funikov S, Burov A, Spasskaya D, Ignatyuk V, Astakhova T, Lyupina Y, Deikin A, Tutyaeva V, Bal N, Karpov V, Morozov A. Immunoproteasome Inhibitor ONX-0914 Affects Long-Term Potentiation in Murine Hippocampus. J Neuroimmune Pharmacol 2021; 16:7-11. [PMID: 33405099 DOI: 10.1007/s11481-020-09973-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/25/2020] [Indexed: 12/29/2022]
Affiliation(s)
- Alexander Maltsev
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerovа 5A, 117485, Moscow, Russia
| | - Sergei Funikov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov street 32, 119991, Moscow, Russia
| | - Alexander Burov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov street 32, 119991, Moscow, Russia
| | - Daria Spasskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov street 32, 119991, Moscow, Russia
| | - Vasilina Ignatyuk
- N. K. Kol'tsov Institute of Developmental Biology, Russian Academy of Sciences, Vavilov street, 26 119334, Moscow, Russia
| | - Tatjana Astakhova
- N. K. Kol'tsov Institute of Developmental Biology, Russian Academy of Sciences, Vavilov street, 26 119334, Moscow, Russia
| | - Yulia Lyupina
- N. K. Kol'tsov Institute of Developmental Biology, Russian Academy of Sciences, Vavilov street, 26 119334, Moscow, Russia
| | - Alexey Deikin
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334, Moscow, Russia
| | - Vera Tutyaeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov street 32, 119991, Moscow, Russia
| | - Natalia Bal
- Institute of Higher Nervous Activity and Neurophysiology, Russian Academy of Sciences, Butlerovа 5A, 117485, Moscow, Russia
| | - Vadim Karpov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov street 32, 119991, Moscow, Russia
| | - Alexey Morozov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilov street 32, 119991, Moscow, Russia.
| |
Collapse
|
25
|
Hubbell GE, Tepe JJ. Natural product scaffolds as inspiration for the design and synthesis of 20S human proteasome inhibitors. RSC Chem Biol 2020; 1:305-332. [PMID: 33791679 PMCID: PMC8009326 DOI: 10.1039/d0cb00111b] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022] Open
Abstract
The 20S proteasome is a valuable target for the treatment of a number of diseases including cancer, neurodegenerative disease, and parasitic infection. In an effort to discover novel inhibitors of the 20S proteasome, many reseaarchers have looked to natural products as potential leads for drug discovery. The following review discusses the efforts made in the field to isolate and identify natural products as inhibitors of the proteasome. In addition, we describe some of the modifications made to natural products in order to discover more potent and selective inhibitors for potential disease treatment.
Collapse
Affiliation(s)
- Grace E. Hubbell
- Department of Chemistry, Michigan State UniversityEast LansingMI 48823USA
| | - Jetze J. Tepe
- Department of Chemistry, Michigan State UniversityEast LansingMI 48823USA
| |
Collapse
|
26
|
Maurits E, Degeling CG, Kisselev AF, Florea BI, Overkleeft HS. Structure-Based Design of Fluorogenic Substrates Selective for Human Proteasome Subunits. Chembiochem 2020; 21:3220-3224. [PMID: 32598532 PMCID: PMC7754458 DOI: 10.1002/cbic.202000375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 06/29/2020] [Indexed: 11/07/2022]
Abstract
Proteasomes are established therapeutic targets for hematological cancers and promising targets for autoimmune diseases. In the past, we have designed and synthesized mechanism-based proteasome inhibitors that are selective for the individual catalytic activities of human constitutive proteasomes and immunoproteasomes: β1c, β1i, β2c, β2i, β5c and β5i. We show here that by taking the oligopeptide recognition element and substituting the electrophile for a fluorogenic leaving group, fluorogenic substrates are obtained that report on the proteasome catalytic activity also targeted by the parent inhibitor. Though not generally applicable (β5c and β2i substrates showing low activity), effective fluorogenic substrates reporting on the individual activity of β1c, β1i, β2c and β5i subunits in Raji (human B cell) lysates and purified 20S proteasome were identified in this manner. Our work thus adds to the expanding proteasome research toolbox through the identification of new and/or more effective subunit-selective fluorogenic substrates.
Collapse
Affiliation(s)
- Elmer Maurits
- Leiden Institute of ChemistryLeiden UniversityEinsteinweg 552333 CCLeidenThe Netherlands
| | - Christian G. Degeling
- Leiden Institute of ChemistryLeiden UniversityEinsteinweg 552333 CCLeidenThe Netherlands
| | - Alexei F. Kisselev
- Department of Drug Discovery and DevelopmentHarrison School of PharmacyAuburn UniversityAuburnAL 36849USA
| | - Bogdan I. Florea
- Leiden Institute of ChemistryLeiden UniversityEinsteinweg 552333 CCLeidenThe Netherlands
| | - Herman S. Overkleeft
- Leiden Institute of ChemistryLeiden UniversityEinsteinweg 552333 CCLeidenThe Netherlands
| |
Collapse
|
27
|
Recent insights how combined inhibition of immuno/proteasome subunits enables therapeutic efficacy. Genes Immun 2020; 21:273-287. [PMID: 32839530 DOI: 10.1038/s41435-020-00109-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/03/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022]
Abstract
The proteasome is a multicatalytic protease in the cytosol and nucleus of all eukaryotic cells that controls numerous cellular processes through regulated protein degradation. Proteasome inhibitors have significantly improved the survival of multiple myeloma patients. However, clinically approved proteasome inhibitors have failed to show efficacy against solid tumors, neither alone nor in combination with other therapies. Targeting the immunoproteasome with selective inhibitors has been therapeutically effective in preclinical models for several autoimmune diseases and colon cancer. Moreover, immunoproteasome inhibitors prevented the chronic rejection of allogeneic organ transplants. In recent years, it has become apparent that inhibition of one single active center of the proteasome is insufficient to achieve therapeutic benefits. In this review we summarize the latest insights how targeting multiple catalytically active proteasome subunits can interfere with disease progression in autoimmunity, growth of solid tumors, and allograft rejection.
Collapse
|
28
|
Maurits E, van de Graaff MJ, Maiorana S, Wander DPA, Dekker PM, van der Zanden SY, Florea BI, Neefjes JJC, Overkleeft HS, van Kasteren SI. Immunoproteasome Inhibitor-Doxorubicin Conjugates Target Multiple Myeloma Cells and Release Doxorubicin upon Low-Dose Photon Irradiation. J Am Chem Soc 2020; 142:7250-7253. [PMID: 32275401 PMCID: PMC7181259 DOI: 10.1021/jacs.9b11969] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
Proteasome
inhibitors are established therapeutic agents for the
treatment of hematological cancers, as are anthracyclines such
as doxorubicin. We here present a new drug targeting approach that
combines both drug classes into a single molecule. Doxorubicin was
conjugated to an immunoproteasome-selective inhibitor via light-cleavable
linkers, yielding peptide epoxyketone–doxorubicin prodrugs
that remained selective and active toward immunoproteasomes.
Upon cellular uptake and immunoproteasome inhibition, doxorubicin
is released from the immunoproteasome inhibitor through photoirradiation.
Multiple myeloma cells in this way take a double hit: immunoproteasome
inhibition and doxorubicin-induced toxicity. Our strategy, which entails
targeting of a cytotoxic agent, through a covalent enzyme inhibitor
that is detrimental to tumor tissue in its own right, may find use
in the search for improved anticancer drugs.
Collapse
Affiliation(s)
- Elmer Maurits
- Gorlaeus Laboratories, Leiden Institute of Chemistry, Leiden University, Leiden 2333 CC, The Netherlands
| | - Michel J van de Graaff
- Gorlaeus Laboratories, Leiden Institute of Chemistry, Leiden University, Leiden 2333 CC, The Netherlands
| | - Santina Maiorana
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98122 Messina, Italy
| | - Dennis P A Wander
- Gorlaeus Laboratories, Leiden Institute of Chemistry, Leiden University, Leiden 2333 CC, The Netherlands
| | - Patrick M Dekker
- Gorlaeus Laboratories, Leiden Institute of Chemistry, Leiden University, Leiden 2333 CC, The Netherlands
| | | | - Bogdan I Florea
- Gorlaeus Laboratories, Leiden Institute of Chemistry, Leiden University, Leiden 2333 CC, The Netherlands
| | - Jacques J C Neefjes
- ONCODE Institute, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Herman S Overkleeft
- Gorlaeus Laboratories, Leiden Institute of Chemistry, Leiden University, Leiden 2333 CC, The Netherlands
| | - Sander I van Kasteren
- Gorlaeus Laboratories, Leiden Institute of Chemistry, Leiden University, Leiden 2333 CC, The Netherlands
| |
Collapse
|
29
|
Bhattarai D, Lee MJ, Baek A, Yeo IJ, Miller Z, Baek YM, Lee S, Kim DE, Hong JT, Kim KB. LMP2 Inhibitors as a Potential Treatment for Alzheimer’s Disease. J Med Chem 2020; 63:3763-3783. [DOI: 10.1021/acs.jmedchem.0c00416] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Deepak Bhattarai
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone, Lexington, Kentucky 40536-0596, United States
| | - Min Jae Lee
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone, Lexington, Kentucky 40536-0596, United States
| | - Ahruem Baek
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - In Jun Yeo
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Zachary Miller
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone, Lexington, Kentucky 40536-0596, United States
| | - Yu Mi Baek
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Sukyeong Lee
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Cheongju, Chungbuk 28160, Republic of Korea
| | - Kyung Bo Kim
- Department of Pharmaceutical Sciences, University of Kentucky, 789 South Limestone, Lexington, Kentucky 40536-0596, United States
| |
Collapse
|
30
|
Wang X, Meul T, Meiners S. Exploring the proteasome system: A novel concept of proteasome inhibition and regulation. Pharmacol Ther 2020; 211:107526. [PMID: 32173559 DOI: 10.1016/j.pharmthera.2020.107526] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 03/08/2020] [Indexed: 12/13/2022]
Abstract
The proteasome is a well-identified therapeutic target for cancer treatment. It acts as the main protein degradation system in the cell and degrades key mediators of cell growth, survival and function. The term "proteasome" embraces a whole family of distinct complexes, which share a common proteolytic core, the 20S proteasome, but differ by their attached proteasome activators. Each of these proteasome complexes plays specific roles in the control of cellular function. In addition, distinct proteasome interacting proteins regulate proteasome activity in subcellular compartments and in response to cellular signals. Proteasome activators and regulators may thus serve as building blocks to fine-tune proteasome function in the cell according to cellular needs. Inhibitors of the proteasome, e.g. the FDA approved drugs Velcade™, Kyprolis™, Ninlaro™, inactivate the catalytic 20S core and effectively block protein degradation of all proteasome complexes in the cell resulting in inhibition of cell growth and induction of apoptosis. Efficacy of these inhibitors, however, is hampered by their pronounced cytotoxic side-effects as well as by the emerging development of resistance to catalytic proteasome inhibitors. Targeted inhibition of distinct buiding blocks of the proteasome system, i.e. proteasome activators or regulators, represents an alternative strategy to overcome these limitations. In this review, we stress the importance of the diversity of the proteasome complexes constituting an entire proteasome system. Our building block concept provides a rationale for the defined targeting of distinct proteasome super-complexes in disease. We thereby aim to stimulate the development of innovative therapeutic approaches beyond broad catalytic proteasome inhibition.
Collapse
Affiliation(s)
- Xinyuan Wang
- Comprehensive Pneumology Center (CPC), University Hospital of the Ludwig-Maximilians-University (LMU) and Helmholtz Zentrum München, German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Thomas Meul
- Comprehensive Pneumology Center (CPC), University Hospital of the Ludwig-Maximilians-University (LMU) and Helmholtz Zentrum München, German Center for Lung Research (DZL), 81377 Munich, Germany
| | - Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital of the Ludwig-Maximilians-University (LMU) and Helmholtz Zentrum München, German Center for Lung Research (DZL), 81377 Munich, Germany.
| |
Collapse
|
31
|
Pawar A, Basler M, Goebel H, Alvarez Salinas GO, Groettrup M, Böttcher T. Competitive Metabolite Profiling of Natural Products Reveals Subunit Specific Inhibitors of the 20S Proteasome. ACS CENTRAL SCIENCE 2020; 6:241-246. [PMID: 32123742 PMCID: PMC7047272 DOI: 10.1021/acscentsci.9b01170] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Indexed: 05/11/2023]
Abstract
We have developed a syringolin-based chemical probe and explored its utility for the profiling of metabolite extracts as potent inhibitors of the 20S proteasome. Activity-guided fractionation by competitive labeling allowed us to isolate and identify glidobactin A and C as well as luminmycin A from a Burkholderiales strain. The natural products exhibited unique subunit specificities for the proteolytic subunits of human and mouse constitutive and immunoproteasome in the lower nanomolar range. In particular, glidobactin C displayed an unprecedented β2/β5 coinhibition profile with single-digit nanomolar potency in combination with sufficiently high cell permeability. These properties render glidobactin C a promising live cell proteasome inhibitor with potent activity against human breast cancer cell lines and comparably low immunotoxicity.
Collapse
Affiliation(s)
- Atul Pawar
- Department
of Chemistry, Zukunftskolleg, Konstanz Research School Chemical Biology, University of Konstanz, 78457 Konstanz, Germany
| | - Michael Basler
- Division
of Immunology, Department of Biology, University
of Konstanz, 78457 Konstanz, Germany
- Biotechnology
Institute Thurgau, 8280 Kreuzlingen, Switzerland
| | - Heike Goebel
- Division
of Immunology, Department of Biology, University
of Konstanz, 78457 Konstanz, Germany
- Biotechnology
Institute Thurgau, 8280 Kreuzlingen, Switzerland
| | - Gerardo Omar Alvarez Salinas
- Division
of Immunology, Department of Biology, University
of Konstanz, 78457 Konstanz, Germany
- Biotechnology
Institute Thurgau, 8280 Kreuzlingen, Switzerland
| | - Marcus Groettrup
- Division
of Immunology, Department of Biology, University
of Konstanz, 78457 Konstanz, Germany
- Biotechnology
Institute Thurgau, 8280 Kreuzlingen, Switzerland
| | - Thomas Böttcher
- Department
of Chemistry, Zukunftskolleg, Konstanz Research School Chemical Biology, University of Konstanz, 78457 Konstanz, Germany
| |
Collapse
|
32
|
|
33
|
Xin BT, Espinal C, de Bruin G, Filippov DV, van der Marel GA, Florea BI, Overkleeft HS. Two-Step Bioorthogonal Activity-Based Protein Profiling of Individual Human Proteasome Catalytic Sites. Chembiochem 2020; 21:248-255. [PMID: 31597011 DOI: 10.1002/cbic.201900551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Indexed: 12/12/2022]
Abstract
Bioorthogonal chemistry allows the selective modification of biomolecules in complex biological samples. One application of this methodology is in two-step activity-based protein profiling (ABPP), a methodology that is particularly attractive where direct ABPP using fluorescent or biotinylated probes is ineffective. Herein we describe a set of norbornene-modified, mechanism-based proteasome inhibitors aimed to be selective for each of the six catalytic sites of human constitutive proteasomes and immunoproteasomes. The probes developed for β1i, β2i, β5c, and β5i proved to be useful two-step ABPs that effectively label their developed proteasome subunits in both Raji cell extracts and living Raji cells through inverse-electron-demand Diels-Alder (iEDDA) ligation. The compound developed for β1c proved incapable of penetrating the cell membrane, but effectively labels β1c in vitro. The compound developed for β2c proved not selective, but its azide-containing analogue LU-002c proved effective in labeling of β2c via azide-alkyne click ligation chemistry both in vitro and in situ. In total, our results contribute to the growing list of proteasome activity tools to include five subunit-selective activity-based proteasome probes, four of which report on proteasome activities in living cells.
Collapse
Affiliation(s)
- Bo-Tao Xin
- Gorlaeus Laboratories, Leiden Institute of Chemistry, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Christofer Espinal
- Gorlaeus Laboratories, Leiden Institute of Chemistry, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Gerjan de Bruin
- Gorlaeus Laboratories, Leiden Institute of Chemistry, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.,Present address, Acerta Pharma B.V., Industrielaan 63, 5349 AE, Oss, The Netherlands
| | - Dmitri V Filippov
- Gorlaeus Laboratories, Leiden Institute of Chemistry, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Gijsbert A van der Marel
- Gorlaeus Laboratories, Leiden Institute of Chemistry, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Bogdan I Florea
- Gorlaeus Laboratories, Leiden Institute of Chemistry, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Herman S Overkleeft
- Gorlaeus Laboratories, Leiden Institute of Chemistry, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| |
Collapse
|
34
|
KONG L, LU J, ZHU H, ZHANG J. [Research progress on selective immunoproteasome inhibitors]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2019; 48:688-694. [PMID: 31955545 PMCID: PMC8800774 DOI: 10.3785/j.issn.1008-9292.2019.12.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/26/2019] [Indexed: 06/10/2023]
Abstract
Immunoproteasome is associated with various diseases such as hematologic malignancies, inflammatory, autoimmune and central nervous system diseases, and over expression of immunoproteasome is observed in all of these diseases. Immunoproteasome inhibitors can reduce the expression of immunoproteasome by inhibiting the production of related cell-inducing factors and the activity of T lymphocyte for treating related diseases. In order to achieve good efficacy and reduce the toxic effects, key for development of selective immunoproteasome inhibitors is the high selectivity and potent activity of the three active subunits of the proteasome. This review summarizes the structure and functions of immunoproteasome and the associated diseases. Besides, structure, activity and status of selective immunoproteasome inhibitors are also been highlighted.
Collapse
Affiliation(s)
| | | | | | - Jiankang ZHANG
- 张建康(1987-), 男, 博士, 讲师, 硕士生导师, 主要从事抗肿瘤药物研发工作, E-mail:
;
https://orcid.org/0000-0003-0365-7238
| |
Collapse
|
35
|
A dual inhibitor of the proteasome catalytic subunits LMP2 and Y attenuates disease progression in mouse models of Alzheimer's disease. Sci Rep 2019; 9:18393. [PMID: 31804556 PMCID: PMC6895163 DOI: 10.1038/s41598-019-54846-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/15/2019] [Indexed: 12/18/2022] Open
Abstract
The immunoproteasome (iP) is a variant of the constitutive proteasome (cP) that is abundantly expressed in immune cells which can also be induced in somatic cells by cytokines such as TNF-α or IFN-γ. Accumulating evidence support that the iP is closely linked to multiple facets of inflammatory response, eventually leading to the development of several iP inhibitors as potential therapeutic agents for autoimmune diseases. Recent studies also found that the iP is upregulated in reactive glial cells surrounding amyloid β (Aβ) deposits in brains of Alzheimer’s disease (AD) patients, but the role it plays in the pathogenesis of AD remains unclear. In this study, we investigated the effects of several proteasome inhibitors on cognitive function in AD mouse models and found that YU102, a dual inhibitor of the iP catalytic subunit LMP2 and the cP catalytic subunit Y, ameliorates cognitive impairments in AD mouse models without affecting Aβ deposition. The data obtained from our investigation revealed that YU102 suppresses the secretion of inflammatory cytokines from microglial cells. Overall, this study indicates that there may exist a potential link between LMP2/Y and microglia-mediated neuroinflammation and that inhibition of these subunits may offer a new therapeutic strategy for AD.
Collapse
|
36
|
Stubba D, Bensinger D, Steinbacher J, Proskurjakov L, Salcedo Gómez Á, Schmidt U, Roth S, Schmitz K, Schmidt B. Cell-Based Optimization of Covalent Reversible Ketoamide Inhibitors Bridging the Unprimed to the Primed Site of the Proteasome β5 Subunit. ChemMedChem 2019; 14:2005-2022. [PMID: 31675179 PMCID: PMC6916368 DOI: 10.1002/cmdc.201900472] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/01/2019] [Indexed: 12/11/2022]
Abstract
The ubiquitin-proteasome system (UPS) is an established therapeutic target for approved drugs to treat selected hematologic malignancies. While drug discovery targeting the UPS focuses on irreversibly binding epoxyketones and slowly-reversibly binding boronates, optimization of novel covalent-reversibly binding warheads remains largely unattended. We previously reported α-ketoamides to be a promising reversible lead motif, yet the cytotoxic activity required further optimization. This work focuses on the lead optimization of phenoxy-substituted α-ketoamides combining the structure-activity relationships from the primed and the non-primed site of the proteasome β5 subunit. Our optimization strategy is accompanied by molecular modeling, suggesting occupation of P1' by a 3-phenoxy group to increase β5 inhibition and cytotoxic activity in leukemia cell lines. Key compounds were further profiled for time-dependent inhibition of cellular substrate conversion. Furthermore, the α-ketoamide lead structure 27 does not affect escape response behavior in Danio rerio embryos, in contrast to bortezomib, which suggests increased target specificity.
Collapse
Affiliation(s)
- Daniel Stubba
- Clemens-Schoepf-Institute for Organic Chemistry & BiochemistryTechnische Universität DarmstadtAlarich-Weiss-Str. 464287DarmstadtGermany
| | - Dennis Bensinger
- Clemens-Schoepf-Institute for Organic Chemistry & BiochemistryTechnische Universität DarmstadtAlarich-Weiss-Str. 464287DarmstadtGermany
| | - Janika Steinbacher
- Clemens-Schoepf-Institute for Organic Chemistry & BiochemistryTechnische Universität DarmstadtAlarich-Weiss-Str. 464287DarmstadtGermany
| | - Lilia Proskurjakov
- Clemens-Schoepf-Institute for Organic Chemistry & BiochemistryTechnische Universität DarmstadtAlarich-Weiss-Str. 464287DarmstadtGermany
| | - Álvaro Salcedo Gómez
- Clemens-Schoepf-Institute for Organic Chemistry & BiochemistryTechnische Universität DarmstadtAlarich-Weiss-Str. 464287DarmstadtGermany
| | - Uwe Schmidt
- Visual Inference Lab, Department of Computer ScienceTechnische Universität DarmstadtHuchschulstr. 1064289DarmstadtGermany
| | - Stefan Roth
- Visual Inference Lab, Department of Computer ScienceTechnische Universität DarmstadtHuchschulstr. 1064289DarmstadtGermany
| | - Katja Schmitz
- Clemens-Schoepf-Institute for Organic Chemistry & BiochemistryTechnische Universität DarmstadtAlarich-Weiss-Str. 464287DarmstadtGermany
| | - Boris Schmidt
- Clemens-Schoepf-Institute for Organic Chemistry & BiochemistryTechnische Universität DarmstadtAlarich-Weiss-Str. 464287DarmstadtGermany
| |
Collapse
|
37
|
Schiffrer ES, Sosič I, Šterman A, Mravljak J, Raščan IM, Gobec S, Gobec M. A focused structure-activity relationship study of psoralen-based immunoproteasome inhibitors. MEDCHEMCOMM 2019; 10:1958-1965. [PMID: 32952997 PMCID: PMC7478164 DOI: 10.1039/c9md00365g] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/08/2019] [Indexed: 12/22/2022]
Abstract
The immunoproteasome is a multicatalytic protease that is predominantly expressed in cells of hematopoietic origin. Its elevated expression has been associated with autoimmune diseases, various types of cancer, and inflammatory diseases. The development of immunoproteasome-selective inhibitors with non-peptidic scaffolds remains a challenging task. Here, we describe a focused series of psoralen-based inhibitors of the β5i subunit of the immunoproteasome with different substituents placed at position 4'. The most promising compound was further evaluated through changes at position 3 of the psoralen ring. Despite a small decrease in the inhibitory potency in comparison with the parent compound, we were able to improve the selectivity against other subunits of both the immunoproteasome and the constitutive proteasome. The most potent compounds discriminated between both proteasome types in cell lysates and also showed a decrease in cytokine secretion in peripheral blood mononuclear cells.
Collapse
Affiliation(s)
- Eva Shannon Schiffrer
- Faculty of Pharmacy , Chair of Pharmaceutical Chemistry , University of Ljubljana , Aškerčeva 7 , 1000 Ljubljana , Slovenia
| | - Izidor Sosič
- Faculty of Pharmacy , Chair of Pharmaceutical Chemistry , University of Ljubljana , Aškerčeva 7 , 1000 Ljubljana , Slovenia
| | - Andrej Šterman
- Faculty of Pharmacy , Chair of Pharmaceutical Chemistry , University of Ljubljana , Aškerčeva 7 , 1000 Ljubljana , Slovenia
| | - Janez Mravljak
- Faculty of Pharmacy , Chair of Pharmaceutical Chemistry , University of Ljubljana , Aškerčeva 7 , 1000 Ljubljana , Slovenia
| | - Irena Mlinarič Raščan
- Faculty of Pharmacy , Chair of Clinical Biochemistry , University of Ljubljana , Aškerčeva 7 , 1000 Ljubljana , Slovenia . ; Tel: +386 1 476 9636
| | - Stanislav Gobec
- Faculty of Pharmacy , Chair of Pharmaceutical Chemistry , University of Ljubljana , Aškerčeva 7 , 1000 Ljubljana , Slovenia
| | - Martina Gobec
- Faculty of Pharmacy , Chair of Clinical Biochemistry , University of Ljubljana , Aškerčeva 7 , 1000 Ljubljana , Slovenia . ; Tel: +386 1 476 9636
| |
Collapse
|
38
|
Xi J, Zhuang R, Kong L, He R, Zhu H, Zhang J. Immunoproteasome-selective inhibitors: An overview of recent developments as potential drugs for hematologic malignancies and autoimmune diseases. Eur J Med Chem 2019; 182:111646. [PMID: 31521028 DOI: 10.1016/j.ejmech.2019.111646] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/24/2019] [Accepted: 08/25/2019] [Indexed: 12/23/2022]
Abstract
The immunoproteasome, a specialized form of proteasome, is mainly expressed in lymphocytes and monocytes of jawed vertebrates and responsible for the generation of antigenic peptides for cell-mediated immunity. Overexpression of immunoproteasome have been detected in a wide range of diseases including malignancies, autoimmune and inflammatory diseases. Following the successful approval of constitutive proteasome inhibitors bortezomib, carfilzomib and Ixazomib, and with the clarification of immunoproteasome crystal structure and functions, a variety of immunoproteasome inhibitors were discovered or rationally developed. Not only the inhibitory activities, the selectivities for immunoproteasome over constitutive proteasome are essential for the clinical potential of these analogues, which has been validated by the clinical evaluation of immunoproteasome-selective inhibitor KZR-616 for the treatment of systemic lupus erythematosus. In this review, structure, function as well as the current developments of various inhibitors against immunoproteasome are going to be summarized, which help to fully understand the target for drug discovery.
Collapse
Affiliation(s)
- Jianjun Xi
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, Zhejiang Province, China
| | - Rangxiao Zhuang
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, Zhejiang Province, China
| | - Limin Kong
- Department of Pharmacy, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310003, Zhejiang Province, China
| | - Ruoyu He
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, Zhejiang Province, China
| | - Huajian Zhu
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, Zhejiang Province, China
| | - Jiankang Zhang
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, Zhejiang Province, China.
| |
Collapse
|
39
|
Allardyce DJ, Bell CM, Loizidou EZ. Argyrin B, a non-competitive inhibitor of the human immunoproteasome exhibiting preference for β1i. Chem Biol Drug Des 2019; 94:1556-1567. [PMID: 31074944 DOI: 10.1111/cbdd.13539] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 03/12/2019] [Accepted: 03/31/2019] [Indexed: 12/14/2022]
Abstract
Inhibitors of the proteasome have found broad therapeutic applications; however, they show severe toxicity due to the abundance of proteasomes in healthy cells. In contrast, inhibitors of the immunoproteasome, which is upregulated during disease states, are less toxic and have increased therapeutic potential including against autoimmune disorders. In this project, we report argyrin B, a natural product cyclic peptide to be a reversible, non-competitive inhibitor of the immunoproteasome. Argyrin B showed selective inhibition of the β5i and β1i sites of the immunoproteasome over the β5c and β1c sites of the constitutive proteasome with nearly 20-fold selective inhibition of β1i over the homologous β1c. Molecular modelling attributes the β1i over β1c selectivity to the small hydrophobic S1 pocket of β1i and β5i over β5c to site-specific amino acid variations that enable additional bonding interactions and stabilization of the binding conformation. These findings facilitate the design of immunoproteasome selective and reversible inhibitors that may have a greater therapeutic potential and lower toxicity.
Collapse
Affiliation(s)
- Duncan J Allardyce
- Department of Natural Sciences, Faculty of Science and Technology, Middlesex University, London, UK
| | - Celia M Bell
- Department of Natural Sciences, Faculty of Science and Technology, Middlesex University, London, UK
| | - Eriketi Z Loizidou
- Department of Natural Sciences, Faculty of Science and Technology, Middlesex University, London, UK
| |
Collapse
|
40
|
Lee MJ, Bhattarai D, Yoo J, Miller Z, Park JE, Lee S, Lee W, Driscoll JJ, Kim KB. Development of Novel Epoxyketone-Based Proteasome Inhibitors as a Strategy To Overcome Cancer Resistance to Carfilzomib and Bortezomib. J Med Chem 2019; 62:4444-4455. [PMID: 30964987 DOI: 10.1021/acs.jmedchem.8b01943] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Over the past 15 years, proteasome inhibitors (PIs), namely bortezomib, carfilzomib (Cfz) and ixazomib, have significantly improved the overall survival and quality-of-life for multiple myeloma (MM) patients. However, a significant portion of MM patients do not respond to PI therapies. Drug resistance is present either de novo or acquired after prolonged therapy through mechanisms that remain poorly defined. The lack of a clear understanding of clinical PI resistance has hampered the development of next-generation PI drugs to treat MM patients who no longer respond to currently available therapies. Here, we designed and synthesized novel epoxyketone-based PIs by structural modifications at the P1' site. We show that a Cfz analog, 9, harboring a hydroxyl substituent at its P1' position was highly cytotoxic against cancer cell lines displaying de novo or acquired resistance to Cfz. These results suggest that peptide epoxyketones incorporating P1'-targeting moieties may have the potential to bypass resistance mechanisms associated with Cfz and to provide additional clinical options for patients resistant to Cfz.
Collapse
Affiliation(s)
- Min Jae Lee
- Department of Pharmaceutical Sciences , University of Kentucky , Lexington , Kentucky 40536 , United States
| | - Deepak Bhattarai
- Department of Pharmaceutical Sciences , University of Kentucky , Lexington , Kentucky 40536 , United States
| | - Jisu Yoo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences , Seoul National University , Seoul 08826 , Korea
| | - Zach Miller
- Department of Pharmaceutical Sciences , University of Kentucky , Lexington , Kentucky 40536 , United States
| | - Ji Eun Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences , Seoul National University , Seoul 08826 , Korea
| | - Sukyeong Lee
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology , Baylor College of Medicine , Houston , Texas 77030 , United States
| | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences , Seoul National University , Seoul 08826 , Korea
| | - James J Driscoll
- Department of Internal Medicine , Division of Hematology and Oncology and University of Cincinnati Cancer Institute , Cincinnati , Ohio 45267 , United States
| | - Kyung Bo Kim
- Department of Pharmaceutical Sciences , University of Kentucky , Lexington , Kentucky 40536 , United States
| |
Collapse
|
41
|
Ettari R, Cerchia C, Maiorana S, Guccione M, Novellino E, Bitto A, Grasso S, Lavecchia A, Zappalà M. Development of Novel Amides as Noncovalent Inhibitors of Immunoproteasomes. ChemMedChem 2019; 14:842-852. [PMID: 30829448 DOI: 10.1002/cmdc.201900028] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 03/04/2019] [Indexed: 01/02/2023]
Abstract
The development of immunoproteasome-selective inhibitors is a promising strategy for treating hematologic malignancies, autoimmune and inflammatory diseases. In this context, we report the design, synthesis, and biological evaluation of a new series of amide derivatives as immunoproteasome inhibitors. Notably, the designed compounds act as noncovalent inhibitors, which might be a promising therapeutic option because of the lack of drawbacks and side effects associated with irreversible inhibition. Among the synthesized compounds, we identified a panel of active inhibitors with Ki values in the low micromolar or sub-micromolar ranges toward the β5i and/or β1i subunits of immunoproteasomes. One of the active compounds was shown to be the most potent and selective inhibitor with a Ki value of 21 nm against the single β1i subunit. Docking studies allowed us to determine the mode of binding of the molecules in the catalytic site of immunoproteasome subunits.
Collapse
Affiliation(s)
- Roberta Ettari
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Annunziata, 98168, Messina, Italy
| | - Carmen Cerchia
- Department of Pharmacy, Drug Discovery Laboratory, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Santina Maiorana
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Annunziata, 98168, Messina, Italy
| | - Manuela Guccione
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Annunziata, 98168, Messina, Italy
| | - Ettore Novellino
- Department of Pharmacy, Drug Discovery Laboratory, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, Via C. Valeria, 98125, Messina, Italy
| | - Silvana Grasso
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Annunziata, 98168, Messina, Italy
| | - Antonio Lavecchia
- Department of Pharmacy, Drug Discovery Laboratory, University of Naples Federico II, Via Domenico Montesano 49, 80131, Naples, Italy
| | - Maria Zappalà
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Annunziata, 98168, Messina, Italy
| |
Collapse
|
42
|
Xin BT, Huber EM, de Bruin G, Heinemeyer W, Maurits E, Espinal C, Du Y, Janssens M, Weyburne ES, Kisselev AF, Florea BI, Driessen C, van der Marel GA, Groll M, Overkleeft HS. Structure-Based Design of Inhibitors Selective for Human Proteasome β2c or β2i Subunits. J Med Chem 2019; 62:1626-1642. [PMID: 30657666 PMCID: PMC6378654 DOI: 10.1021/acs.jmedchem.8b01884] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
Subunit-selective
proteasome inhibitors are valuable tools to assess
the biological and medicinal relevance of individual proteasome active
sites. Whereas the inhibitors for the β1c, β1i, β5c,
and β5i subunits exploit the differences in the substrate-binding
channels identified by X-ray crystallography, compounds selectively
targeting β2c or β2i could not yet be rationally designed
because of the high structural similarity of these two subunits. Here,
we report the development, chemical synthesis, and biological screening
of a compound library that led to the identification of the β2c-
and β2i-selective compounds LU-002c (4; IC50 β2c: 8 nM, IC50 β2i/β2c: 40-fold)
and LU-002i (5; IC50 β2i: 220 nM, IC50 β2c/β2i: 45-fold), respectively. Co-crystal
structures with β2 humanized yeast proteasomes visualize protein–ligand
interactions crucial for subunit specificity. Altogether, organic
syntheses, activity-based protein profiling, yeast mutagenesis, and
structural biology allowed us to decipher significant differences
of β2 substrate-binding channels and to complete the set of
subunit-selective proteasome inhibitors.
Collapse
Affiliation(s)
- Bo-Tao Xin
- Gorlaeus Laboratories , Leiden Institute of Chemistry and Netherlands Proteomics Centre , Einsteinweg 55 , 2333 CC Leiden , Netherlands
| | - Eva M Huber
- Center for Integrated Protein Science at the Department Chemie, Lehrstuhl für Biochemie , Technische Universität München , 85748 Garching , Germany
| | - Gerjan de Bruin
- Gorlaeus Laboratories , Leiden Institute of Chemistry and Netherlands Proteomics Centre , Einsteinweg 55 , 2333 CC Leiden , Netherlands
| | - Wolfgang Heinemeyer
- Center for Integrated Protein Science at the Department Chemie, Lehrstuhl für Biochemie , Technische Universität München , 85748 Garching , Germany
| | - Elmer Maurits
- Gorlaeus Laboratories , Leiden Institute of Chemistry and Netherlands Proteomics Centre , Einsteinweg 55 , 2333 CC Leiden , Netherlands
| | - Christofer Espinal
- Gorlaeus Laboratories , Leiden Institute of Chemistry and Netherlands Proteomics Centre , Einsteinweg 55 , 2333 CC Leiden , Netherlands
| | - Yimeng Du
- Gorlaeus Laboratories , Leiden Institute of Chemistry and Netherlands Proteomics Centre , Einsteinweg 55 , 2333 CC Leiden , Netherlands
| | - Marissa Janssens
- Gorlaeus Laboratories , Leiden Institute of Chemistry and Netherlands Proteomics Centre , Einsteinweg 55 , 2333 CC Leiden , Netherlands
| | - Emily S Weyburne
- Department of Molecular and Systems Biology and Norris Cotton Cancer Center , Geisel School of Medicine at Dartmouth , 1 Medical Centre Drive HB7936 , Lebanon , New Hampshire 03756 , United States
| | - Alexei F Kisselev
- Department of Molecular and Systems Biology and Norris Cotton Cancer Center , Geisel School of Medicine at Dartmouth , 1 Medical Centre Drive HB7936 , Lebanon , New Hampshire 03756 , United States
| | - Bogdan I Florea
- Gorlaeus Laboratories , Leiden Institute of Chemistry and Netherlands Proteomics Centre , Einsteinweg 55 , 2333 CC Leiden , Netherlands
| | - Christoph Driessen
- Department of Hematology and Oncology , Kantonsspital St. Gallen , 9007 St. Gallen , Switzerland
| | - Gijsbert A van der Marel
- Gorlaeus Laboratories , Leiden Institute of Chemistry and Netherlands Proteomics Centre , Einsteinweg 55 , 2333 CC Leiden , Netherlands
| | - Michael Groll
- Center for Integrated Protein Science at the Department Chemie, Lehrstuhl für Biochemie , Technische Universität München , 85748 Garching , Germany
| | - Herman S Overkleeft
- Gorlaeus Laboratories , Leiden Institute of Chemistry and Netherlands Proteomics Centre , Einsteinweg 55 , 2333 CC Leiden , Netherlands
| |
Collapse
|
43
|
Proteasome Inhibition in Multiple Myeloma: Head-to-Head Comparison of Currently Available Proteasome Inhibitors. Cell Chem Biol 2019; 26:340-351.e3. [PMID: 30612952 DOI: 10.1016/j.chembiol.2018.11.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/18/2018] [Accepted: 11/12/2018] [Indexed: 12/30/2022]
Abstract
Proteasome inhibitors (PIs) are a backbone of multiple myeloma (MM) therapy. The proteasome harbors six proteolytically active subunits (β1, β2, β5), while β5 was identified as rate-limiting and is a primary target of clinically available PIs. The most effective pattern of subunit inhibition provided by these PIs for cytotoxic activity in MM is unknown. A head-to-head comparison of clinically available PIs shows that in the clinically relevant setting only the co-inhibition of β1 or β2 with β5 activity achieves meaningful functional proteasome inhibition and cytotoxicity, while the selective β2/β5 inhibition of both constitutive and immunoproteasome is the most cytotoxic. In the long-term setting, selective inhibition of β5 subunit is sufficient to induce cytotoxicity in PI-sensitive, but not in PI-resistant MM, and the β5/β2 co-inhibition is the most cytotoxic in PI-resistant MM. These results give a rational basis for selecting individual PIs for the treatment of MM.
Collapse
|
44
|
Johnson HWB, Lowe E, Anderl JL, Fan A, Muchamuel T, Bowers S, Moebius DC, Kirk C, McMinn DL. Required Immunoproteasome Subunit Inhibition Profile for Anti-Inflammatory Efficacy and Clinical Candidate KZR-616 ((2 S,3 R)- N-(( S)-3-(Cyclopent-1-en-1-yl)-1-(( R)-2-methyloxiran-2-yl)-1-oxopropan-2-yl)-3-hydroxy-3-(4-methoxyphenyl)-2-(( S)-2-(2-morpholinoacetamido)propanamido)propenamide). J Med Chem 2018; 61:11127-11143. [PMID: 30380863 DOI: 10.1021/acs.jmedchem.8b01201] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Selective immunoproteasome inhibition is a promising approach for treating autoimmune disorders, but optimal proteolytic active site subunit inhibition profiles remain unknown. We reveal here our design of peptide epoxyketone-based selective low molecular mass polypeptide-7 (LMP7) and multicatalytic endopeptidase complex subunit-1 (MECL-1) subunit inhibitors. Utilizing these and our previously disclosed low molecular mass polypeptide-2 (LMP2) inhibitor, we demonstrate a requirement of dual LMP7/LMP2 or LMP7/MECL-1 subunit inhibition profiles for potent cytokine expression inhibition and in vivo efficacy in an inflammatory disease model. These and additional findings toward optimized solubility led the design and selection of KZR-616 disclosed here and presently in clinical trials for treatment of rheumatic disease.
Collapse
Affiliation(s)
- Henry W B Johnson
- Kezar Life Sciences , 4000 Shoreline Court, Suite 300 , South San Francisco , California 94080 , United States
| | - Eric Lowe
- Kezar Life Sciences , 4000 Shoreline Court, Suite 300 , South San Francisco , California 94080 , United States
| | - Janet L Anderl
- Kezar Life Sciences , 4000 Shoreline Court, Suite 300 , South San Francisco , California 94080 , United States
| | - Andrea Fan
- Kezar Life Sciences , 4000 Shoreline Court, Suite 300 , South San Francisco , California 94080 , United States
| | - Tony Muchamuel
- Kezar Life Sciences , 4000 Shoreline Court, Suite 300 , South San Francisco , California 94080 , United States
| | - Simeon Bowers
- Onyx Pharmaceuticals, an Amgen Subsidiary , South San Francisco , California 94080 , United States
| | - David C Moebius
- Onyx Pharmaceuticals, an Amgen Subsidiary , South San Francisco , California 94080 , United States
| | - Christopher Kirk
- Kezar Life Sciences , 4000 Shoreline Court, Suite 300 , South San Francisco , California 94080 , United States
| | - Dustin L McMinn
- Kezar Life Sciences , 4000 Shoreline Court, Suite 300 , South San Francisco , California 94080 , United States
| |
Collapse
|
45
|
Basler M, Lindstrom MM, LaStant JJ, Bradshaw JM, Owens TD, Schmidt C, Maurits E, Tsu C, Overkleeft HS, Kirk CJ, Langrish CL, Groettrup M. Co-inhibition of immunoproteasome subunits LMP2 and LMP7 is required to block autoimmunity. EMBO Rep 2018; 19:e46512. [PMID: 30279279 PMCID: PMC6280796 DOI: 10.15252/embr.201846512] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 09/04/2018] [Accepted: 09/10/2018] [Indexed: 12/11/2022] Open
Abstract
Cells of hematopoietic origin express high levels of the immunoproteasome, a cytokine-inducible proteasome variant comprising the proteolytic subunits LMP2 (β1i), MECL-1 (β2i), and LMP7 (β5i). Targeting the immunoproteasome in pre-clinical models of autoimmune diseases with the epoxyketone inhibitor ONX 0914 has proven to be effective. ONX 0914 was previously described as a selective LMP7 inhibitor. Here, we show that PRN1126, developed as an exclusively LMP7-specific inhibitor, has limited effects on IL-6 secretion, experimental colitis, and experimental autoimmune encephalomyelitis (EAE). We demonstrate that prolonged exposure of cells with ONX 0914 leads to inhibition of both LMP7 and LMP2. Co-inhibition of LMP7 and LMP2 with PRN1126 and LMP2 inhibitors LU-001i or ML604440 impairs MHC class I cell surface expression, IL-6 secretion, and differentiation of naïve T helper cells to T helper 17 cells, and strongly ameliorates disease in experimental colitis and EAE. Hence, co-inhibition of LMP2 and LMP7 appears to be synergistic and advantageous for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Michael Basler
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany
| | | | | | | | | | - Christian Schmidt
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany
- Konstanz Research School Chemical Biology (KoRS-CB), University of Konstanz, Konstanz, Germany
| | - Elmer Maurits
- Leiden Institute of Chemistry, Leiden University, Leiden, The Netherlands
| | - Christopher Tsu
- Takeda Pharmaceuticals International Co., Cambridge, MA, USA
| | | | | | | | - Marcus Groettrup
- Biotechnology Institute Thurgau (BITg) at the University of Konstanz, Kreuzlingen, Switzerland
- Division of Immunology, Department of Biology, University of Konstanz, Konstanz, Germany
| |
Collapse
|
46
|
Lubos M, Dębowski D, Barcińska E, Meid A, Inkielewicz‐Stepniak I, Burster T, Rolka K. Inhibition of human constitutive 20S proteasome and 20S immunoproteasome with novel
N
‐terminally modified peptide aldehydes and their antitumor activity. Pept Sci (Hoboken) 2018. [DOI: 10.1002/pep2.24100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Marta Lubos
- Department of Molecular Biochemistry, Faculty of ChemistryUniversity of Gdansk Gdansk Poland
| | - Dawid Dębowski
- Department of Molecular Biochemistry, Faculty of ChemistryUniversity of Gdansk Gdansk Poland
| | - Ewelina Barcińska
- Department of Medical ChemistryMedical University of Gdansk Gdansk Poland
| | - Annika Meid
- Department of Neurosurgery, Surgery CenterUlm University Medical Center Ulm Germany
| | | | - Timo Burster
- Department of BiologySchool of Science and Technology, Nazarbayev University Astana Kazakhstan Republic
| | - Krzysztof Rolka
- Department of Molecular Biochemistry, Faculty of ChemistryUniversity of Gdansk Gdansk Poland
| |
Collapse
|
47
|
Design, synthesis, and evaluation of cystargolide-based β-lactones as potent proteasome inhibitors. Eur J Med Chem 2018; 157:962-977. [PMID: 30165344 DOI: 10.1016/j.ejmech.2018.08.052] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/15/2018] [Accepted: 08/18/2018] [Indexed: 11/20/2022]
Abstract
The peptidic β-lactone proteasome inhibitors (PIs) cystargolides A and B were used to conduct structure-activity relationship (SAR) studies in order to assess their anticancer potential. A total of 24 different analogs were designed, synthesized and evaluated for proteasome inhibition, for cytotoxicity towards several cancer cell lines, and for their ability to enter intact cells. X-ray crystallographic analysis and subunit selectivity was used to determine the specific subunit binding associated with the structural modification of the β-lactone (P1), peptidic core, (Px and Py), and end-cap (Pz) of our scaffold. The cystargolide derivative 5k, structurally unique at both Py and P1, exhibited the most promising inhibitory activity for the β5 subunit of human proteasomes (IC50 = 3.1 nM) and significant cytotoxicity towards MCF-7 (IC50 = 416 nM), MDA-MB-231 (IC50 = 74 nM) and RPMI 8226 (IC50 = 41 nM) cancer cell lines. Cellular infiltration assays revealed that minor structural modifications have significant effects on the ability of our PIs to inhibit intracellular proteasomes, and we identified 5k as a promising candidate for continued therapeutic studies. Our novel drug lead 5k is a more potent proteasome inhibitor than carfilzomib with mid-to-low nanomolar IC50 measurements and it is cytotoxic against multiple cancer cell lines at levels approaching those of carfilzomib.
Collapse
|
48
|
Dubiella C, Cui H, Groll M. Tunable Probes with Direct Fluorescence Signals for the Constitutive and Immunoproteasome. Angew Chem Int Ed Engl 2018; 55:13330-13334. [PMID: 27709817 DOI: 10.1002/anie.201605753] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/20/2016] [Indexed: 01/24/2023]
Abstract
Electrophiles are commonly used for the inhibition of proteases. Notably, inhibitors of the proteasome, a central determinant of cellular survival and a target of several FDA-approved drugs, are mainly characterized by the reactivity of their electrophilic head groups. We aimed to tune the inhibitory strength of peptidic sulfonate esters by varying the leaving groups. Indeed, proteasome inhibition correlated well with the pKa of the leaving group. The use of fluorophores as leaving groups enabled us to design probes that release a stoichiometric fluorescence signal upon reaction, thereby directly linking proteasome inactivation to the readout. This principle could be applicable to other sulfonyl fluoride based inhibitors and allows the design of sensitive probes for enzymatic studies.
Collapse
Affiliation(s)
- Christian Dubiella
- Center for Integrated Protein Science Munich (CIPSM), Department of Chemistry, Technische Universität München, Lichtenbergstrasse 4, 85748, Garching, Germany.
| | - Haissi Cui
- Center for Integrated Protein Science Munich (CIPSM), Department of Chemistry, Technische Universität München, Lichtenbergstrasse 4, 85748, Garching, Germany
| | - Michael Groll
- Center for Integrated Protein Science Munich (CIPSM), Department of Chemistry, Technische Universität München, Lichtenbergstrasse 4, 85748, Garching, Germany.
| |
Collapse
|
49
|
Meiners S, Evankovich J, Mallampalli RK. The ubiquitin proteasome system as a potential therapeutic target for systemic sclerosis. Transl Res 2018; 198:17-28. [PMID: 29702079 DOI: 10.1016/j.trsl.2018.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/20/2018] [Accepted: 03/24/2018] [Indexed: 01/16/2023]
Abstract
The present review aims to summarize available knowledge on the role of the ubiquitin-proteasome system (UPS) in the pathogenesis of scleroderma and scleroderma-related disease mechanisms. This will provide the reader with a more mechanistic understanding of disease pathogenesis and help to identify putative novel targets within the UPS for potential therapeutic intervention. Because of the heterogenous manifestations of scleroderma, we will primarily focus on conserved mechanisms that are involved in the development of lung scleroderma phenotypes.
Collapse
Affiliation(s)
- Silke Meiners
- Comprehensive Pneumology Center (CPC), University Hospital, Ludwig Maximilians University, Helmholtz Zentrum München, Germany; Comprehensive Pneumology Center, Munich (CPC-M), Germany; Member of the German Center for Lung Research (DZL), Munich, Germany.
| | - John Evankovich
- Pulmonary, Allergy, and Critical Care Medicine, Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rama K Mallampalli
- Pulmonary, Allergy, and Critical Care Medicine, Acute Lung Injury Center of Excellence, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Cell Biology and Physiology, University of Pittsburgh, Pittsburgh, PA, USA; Medical Specialty Service Line, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA.
| |
Collapse
|
50
|
Rut W, Poręba M, Kasperkiewicz P, Snipas SJ, Drąg M. Selective Substrates and Activity-Based Probes for Imaging of the Human Constitutive 20S Proteasome in Cells and Blood Samples. J Med Chem 2018; 61:5222-5234. [DOI: 10.1021/acs.jmedchem.8b00026] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Wioletta Rut
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland
| | - Marcin Poręba
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland
- Program in Cell Death and Survival Networks, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Paulina Kasperkiewicz
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland
- Program in Cell Death and Survival Networks, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Scott J. Snipas
- Program in Cell Death and Survival Networks, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037, United States
| | - Marcin Drąg
- Department of Bioorganic Chemistry, Faculty of Chemistry, Wroclaw University of Science and Technology, Wyb. Wyspianskiego 27, 50-370 Wroclaw, Poland
| |
Collapse
|