1
|
Zattoni J, Vottero P, Carena G, Uliveto C, Pozzati G, Morabito B, Gitari E, Tuszynski J, Aminpour M. A comprehensive primer and review of PROTACs and their In Silico design. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 264:108687. [PMID: 40058081 DOI: 10.1016/j.cmpb.2025.108687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/28/2025] [Accepted: 02/25/2025] [Indexed: 04/05/2025]
Abstract
The cutting-edge technique of Proteolysis Targeting Chimeras, or PROTACs, has gained significant attention as a viable approach for specific protein degradation. This innovative technology has vast potential in fields such as cancer therapy and drug development. The development of effective and specific therapies for a range of diseases is within reach with PROTACs, which can target previously "undruggable" proteins while circumventing the off-target effects of conventional small molecule inhibitors. This manuscript aims to discuss the application of in silico techniques to the design of these groundbreaking molecules and develop PROTAC complexes, in order to identify potential PROTAC candidates with favorable drug-like properties. Additionally, this manuscript reviews the strengths and weaknesses of these methods to demonstrate their utility and highlights the challenges and future prospects of in silico PROTAC design. The present review provides a valuable and beginner-friendly resource for researchers and drug developers interested in using in silico methods for PROTAC design, specifically ternary structure prediction.
Collapse
Affiliation(s)
- Jacopo Zattoni
- Department of Biomedical Engineering, University of Alberta, Edmonton, T6G 1Z2, Canada
| | - Paola Vottero
- Department of Biomedical Engineering, University of Alberta, Edmonton, T6G 1Z2, Canada
| | - Gea Carena
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Chiara Uliveto
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Giulia Pozzati
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Benedetta Morabito
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Ebenezea Gitari
- Department of Biochemistry, University of Alberta, Edmonton, T6G 1Z2, Canada
| | - Jack Tuszynski
- DIMEAS, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy; Department of Physics, University of Alberta, 11335 Saskatchewan Dr NW, Edmonton, T6G 2M9, Canada
| | - Maral Aminpour
- Department of Biomedical Engineering, University of Alberta, Edmonton, T6G 1Z2, Canada.
| |
Collapse
|
2
|
Sülzen H, Fajtova P, O’Donoghue AJ, Silhan J, Boura E. Structural Insights into Salinosporamide a Mediated Inhibition of the Human 20S Proteasome. Molecules 2025; 30:1386. [PMID: 40142161 PMCID: PMC11946101 DOI: 10.3390/molecules30061386] [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: 02/10/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
The 20S proteasome, a critical component of the ubiquitin-proteasome system, plays a central role in regulating protein degradation in eukaryotic cells. Marizomib (MZB), also known as salinosporamide A, is a natural γ-lactam-β-lactone compound derived from Salinispora tropica and is a potent 20S proteasome covalent inhibitor with demonstrated anticancer properties. Its broad-spectrum inhibition of all three proteasome subunits and its ability to cross the blood-brain barrier has made it a promising therapeutic candidate for glioblastoma. In addition to this, MZB also demonstrates significant inhibition against the 20S proteasome of Trichomonas vaginalis (Tv20S), a protozoan parasite, suggesting its potential for parasitic treatments. Here, we present the cryo-EM structure of the human 20S proteasome in complex with MZB at 2.55 Å resolution. This structure reveals the binding mode of MZB to all six catalytic subunits within the two β-rings of the 20S proteasome, providing a detailed molecular understanding of its irreversible inhibitory mechanism. These findings enhance the therapeutic potential of MZB for both cancer and parasitic diseases at the molecular level and highlight marine-derived natural products in targeting the proteasome for therapeutic applications.
Collapse
Affiliation(s)
- Hagen Sülzen
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo namesti 2, 16610 Prague, Czech Republic (P.F.)
| | - Pavla Fajtova
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo namesti 2, 16610 Prague, Czech Republic (P.F.)
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA;
| | - Anthony J. O’Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA;
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Jan Silhan
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo namesti 2, 16610 Prague, Czech Republic (P.F.)
| | - Evzen Boura
- Institute of Organic Chemistry and Biochemistry, Czech Academy of Sciences, Flemingovo namesti 2, 16610 Prague, Czech Republic (P.F.)
| |
Collapse
|
3
|
İnci A, Dökmeci S. Extracellular chaperones in lysosomal storage diseases. Mol Genet Metab 2025; 145:109086. [PMID: 40106871 DOI: 10.1016/j.ymgme.2025.109086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/23/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Lysosomal storage disorders (LSDs) are a diverse group of inherited metabolic disorders characterized by the accumulation of undegraded substrates within lysosomes due to defective lysosomal function. Recent research has highlighted the pivotal role of extracellular chaperones in the pathophysiology of LSDs, revealing their crucial involvement in modulating disease progression. These chaperones aid in stabilizing and refolding misfolded lysosomal enzymes, enhancing their proper trafficking and function, which in turn reduces substrate accumulation. Furthermore, extracellular chaperones have emerged as promising biomarkers, with their levels in bodily fluids offering potential for disease diagnosis and monitoring. This review explores the current understanding of extracellular chaperones in the context of LSDs, examining their mechanisms of action, biomarker and therapeutic potential, and future directions in clinical application of LSDs.
Collapse
Affiliation(s)
- Aslı İnci
- Gazi University School of Medicine, Department of Pediatric Metabolism, Ankara, Turkey; Hacettepe University School of Medicine, Department of Medical Biology, Ankara, Turkey.
| | - Serap Dökmeci
- Hacettepe University School of Medicine, Department of Medical Biology, Ankara, Turkey
| |
Collapse
|
4
|
Hernández JS, Atienzar P, Shamshurin M, Benassi E, Sokolov MN, Feliz M. Luminescence and Generation of Reactive Oxygen Species in Solution by Ta 6Br 12 Clusters. Chemistry 2025; 31:e202403384. [PMID: 39676056 DOI: 10.1002/chem.202403384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 12/17/2024]
Abstract
An intrinsic metal cluster NIR-II emission of the {Ta6Br12}2+ aqua/hydroxocomplexes was determined in aqueous solutions under inert atmosphere. The photoluminescence (PL) is enhanced in D2O, and the lifetime scale expands from nanoseconds to microseconds. Possible cluster emission transitions have been assigned and analyzed from a computational perspective. In the presence of O2, the cluster compound kept its robustness but showed partial quenching of the PL. Stern-Volmer and laser-flash photolysis studies confirmed that quenching is mainly associated with O2 diffusion. Laser-flash photolysis experiments showed that singlet oxygen (1O2) was not detected under measurement conditions. Generation of peroxide and superoxide radical species after irradiation in D2O was confirmed by using luminol as a probe, whereas no hydroxide radical species were detected as evidenced by the emission of the 3-coumarin carboxylic acid (3-CCA) molecular sensor.
Collapse
Affiliation(s)
- Jhon Sebastián Hernández
- Instituto de Tecnología Química, Universitat Politècnica de València - Agencia Estatal Consejo Superior de Investigaciones Científicas, Avd. de los Naranjos s/n, 46022, Valencia, Spain
| | - Pedro Atienzar
- Instituto de Tecnología Química, Universitat Politècnica de València - Agencia Estatal Consejo Superior de Investigaciones Científicas, Avd. de los Naranjos s/n, 46022, Valencia, Spain
| | - Maxim Shamshurin
- Nikolaev Institute of Inorganic Chemistry, Siberian Branch of Russian Academy of Sciences, 3 Akad. Lavrentiev Ave., 630090, Novosibirsk, Russian Federation
| | - Enrico Benassi
- Novosibirsk State University, 2 Pirogov Str., 630090, Novosibirsk, Russian Federation
- Present address: Dept. of Physics, Informatics and Mathematics, University of Modena and Reggio Emilia, 213/B Giuseppe Campi Str., 41125, Modena, Italy
| | - Maxim N Sokolov
- Nikolaev Institute of Inorganic Chemistry, Siberian Branch of Russian Academy of Sciences, 3 Akad. Lavrentiev Ave., 630090, Novosibirsk, Russian Federation
| | - Marta Feliz
- Instituto de Tecnología Química, Universitat Politècnica de València - Agencia Estatal Consejo Superior de Investigaciones Científicas, Avd. de los Naranjos s/n, 46022, Valencia, Spain
| |
Collapse
|
5
|
Sülzen H, Fajtova P, O’Donoghue AJ, Boura E, Silhan J. Structural insights into Salinosporamide A mediated inhibition of the human 20S proteasome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.28.635221. [PMID: 39974992 PMCID: PMC11838377 DOI: 10.1101/2025.01.28.635221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The 20S proteasome, a critical component of the ubiquitin-proteasome system, plays a central role in regulating protein degradation in eukaryotic cells. Marizomib (MZB), a natural γ-lactam-β-lactone compound derived from Salinispora tropica, is a potent 20S proteasome covalent inhibitor with demonstrated anticancer properties. Its broad-spectrum inhibition of all three proteasome subunits and ability to cross the blood-brain barrier has made it a promising therapeutic candidate for glioblastoma. Here, we present the cryo-EM structure of the human 20S proteasome in complex with MZB at 2.55 Å resolution. This structure reveals the binding mode of MZB to all six catalytic subunits within the two β-rings of the 20S proteasome, providing a detailed molecular understanding of its irreversible inhibitory mechanism. These findings explain the therapeutic potential of MZB at the molecular level and highlight marine-derived natural products in targeting the proteasome for anticancer treatment.
Collapse
Affiliation(s)
- Hagen Sülzen
- Institute of Organic Chemistry and Biochemistry AS CR, v.v.i., Flemingovo nam. 2., 166 10 Prague 6, Czech Republic
| | - Pavla Fajtova
- Institute of Organic Chemistry and Biochemistry AS CR, v.v.i., Flemingovo nam. 2., 166 10 Prague 6, Czech Republic
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92037, USA
| | - Anthony J O’Donoghue
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92037, USA
- Center for Discovery and Innovation in Parasitic Diseases, Skaggs School of Pharmacy and pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Dr., La Jolla, CA 92093, USA
| | - Evzen Boura
- Institute of Organic Chemistry and Biochemistry AS CR, v.v.i., Flemingovo nam. 2., 166 10 Prague 6, Czech Republic
| | - Jan Silhan
- Institute of Organic Chemistry and Biochemistry AS CR, v.v.i., Flemingovo nam. 2., 166 10 Prague 6, Czech Republic
| |
Collapse
|
6
|
Balzarini M, Tong J, Gui W, Jayalath IM, Schell BB, Kodadek T. Recruitment to the Proteasome Is Necessary but Not Sufficient for Chemically Induced, Ubiquitin-Independent Degradation of Native Proteins. ACS Chem Biol 2024; 19:2323-2335. [PMID: 39439063 PMCID: PMC11707830 DOI: 10.1021/acschembio.4c00422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Targeted protein degradation (TPD) is a promising strategy for drug development. Most degraders function by forcing the association of the target protein (TP) with an E3 Ubiquitin (Ub) ligase, which, in favorable cases, results in the polyubiquitylation of the TP and its subsequent degradation by the 26S proteasome. An alternative strategy would be to create chemical dimerizers that bypass the requirement for polyubiquitylation by recruiting the target protein directly to the proteasome. Direct-to-proteasome degraders (DPDs) may exhibit different characteristics than ubiquitin-dependent degraders, but few studies of this type of TPD have been published, largely due to the dearth of suitable proteasome ligands. To facilitate studies of DPDs, we report here a mammalian cell line in which the HaloTag protein is fused to the proteasome via Rpn13, one of the ubiquitin receptors. In these cells, a chloroalkane serves as a covalent proteasome ligand surrogate. We show that chimeric molecules comprised of a chloroalkane linked to a ligand for the BET family of proteins or the Cdk2/7/9 family of kinases result in ubiquitin-independent degradation of some of these target proteins. We use this system, the first that allows facile degradation of native proteins in a ubiquitin-independent fashion, to probe two issues: the effect of varying the length of the linker connecting the chloroalkane and the target ligand and the selectivity of degradation within the protein families engaged by the target ligand.
Collapse
Affiliation(s)
- Madeline Balzarini
- Skaggs Graduate School of Chemical and Biological Sciences, 120 Scripps Way, Jupiter, FL 33458. USA
- Department of Chemistry, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 120 Scripps Way, Jupiter, FL 33458. USA
| | - Joel Tong
- Skaggs Graduate School of Chemical and Biological Sciences, 120 Scripps Way, Jupiter, FL 33458. USA
- Department of Chemistry, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 120 Scripps Way, Jupiter, FL 33458. USA
| | - Weijun Gui
- Department of Chemistry, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 120 Scripps Way, Jupiter, FL 33458. USA
| | - Isuru M. Jayalath
- Department of Chemistry, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 120 Scripps Way, Jupiter, FL 33458. USA
| | - Bin-Bin Schell
- Skaggs Graduate School of Chemical and Biological Sciences, 120 Scripps Way, Jupiter, FL 33458. USA
- Department of Chemistry, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 120 Scripps Way, Jupiter, FL 33458. USA
| | - Thomas Kodadek
- Skaggs Graduate School of Chemical and Biological Sciences, 120 Scripps Way, Jupiter, FL 33458. USA
- Department of Chemistry, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, 120 Scripps Way, Jupiter, FL 33458. USA
| |
Collapse
|
7
|
Zhang R, Shen Y, Li X. Tilt-series-based joint CTF estimation for cryo-electron tomography. Structure 2024; 32:1239-1247.e3. [PMID: 38823380 DOI: 10.1016/j.str.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/31/2024] [Accepted: 05/07/2024] [Indexed: 06/03/2024]
Abstract
Contrast transfer function (CTF) estimation is a necessary step in the cryo-electron tomography (cryoET) workflow and essential for high-resolution in situ structural determination. However, the low signal-to-noise ratio and continuous defocus variation in micrographs of cryoET tilt series make accurate CTF estimation challenging. Here, we report a tilt-series-based joint CTF estimation method implemented in the new software CTFMeasure. The joint estimation method combines all Thon-ring signals in a tilt series to improve the estimation accuracy. By using an objective function involving the CTF parameters and geometric parameters of a cryoET tilt series, CTFMeasure can estimate the CTF parameters of each micrograph and the absolute tilt angle offset of the lamellar sample relative to the sample stage plane, which is usually the glancing angle used during focused ion beam (FIB) milling. Tests on both synthetic and experimental data, as well as subtomogram averaging, demonstrated the accurate CTF estimation of cryoET tilt series by CTFMeasure.
Collapse
Affiliation(s)
- Ranhao Zhang
- Key Laboratory for Protein Sciences of Ministry of Education, School of Life Sciences, Tsinghua University, Beijing 100084, China; State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Joint Center for Life Sciences, Beijing 100084, China; Beijing Frontier Research Center for Biological Structure, Beijing 100084, China; School of Life Sciences, Tsinghua University, Beijing 100084, China; Department of Electronic Engineering, Tsinghua University, Beijing 100084, China; Beijing National Research Center for Information Science and Technology, Tsinghua University, Beijing 100084, China
| | - Yuan Shen
- Department of Electronic Engineering, Tsinghua University, Beijing 100084, China; Beijing National Research Center for Information Science and Technology, Tsinghua University, Beijing 100084, China.
| | - Xueming Li
- Key Laboratory for Protein Sciences of Ministry of Education, School of Life Sciences, Tsinghua University, Beijing 100084, China; State Key Laboratory of Membrane Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Joint Center for Life Sciences, Beijing 100084, China; Beijing Frontier Research Center for Biological Structure, Beijing 100084, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
8
|
Adolf F, Du J, Goodall EA, Walsh RM, Rawson S, von Gronau S, Harper JW, Hanna J, Schulman BA. Visualizing chaperone-mediated multistep assembly of the human 20S proteasome. Nat Struct Mol Biol 2024; 31:1176-1188. [PMID: 38600324 PMCID: PMC11327110 DOI: 10.1038/s41594-024-01268-9] [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: 11/05/2023] [Accepted: 03/06/2024] [Indexed: 04/12/2024]
Abstract
Dedicated assembly factors orchestrate the stepwise production of many molecular machines, including the 28-subunit proteasome core particle (CP) that mediates protein degradation. Here we report cryo-electron microscopy reconstructions of seven recombinant human subcomplexes that visualize all five chaperones and the three active site propeptides across a wide swath of the assembly pathway. Comparison of these chaperone-bound intermediates and a matching mature CP reveals molecular mechanisms determining the order of successive subunit additions, as well as how proteasome subcomplexes and assembly factors structurally adapt upon progressive subunit incorporation to stabilize intermediates, facilitate the formation of subsequent intermediates and ultimately rearrange to coordinate proteolytic activation with gated access to active sites. This work establishes a methodologic approach for structural analysis of multiprotein complex assembly intermediates, illuminates specific functions of assembly factors and reveals conceptual principles underlying human proteasome biogenesis, thus providing an explanation for many previous biochemical and genetic observations.
Collapse
Affiliation(s)
- Frank Adolf
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Jiale Du
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Ellen A Goodall
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Richard M Walsh
- Harvard Cryo-Electron Microscopy Center for Structural Biology, Harvard Medical School, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Shaun Rawson
- Harvard Cryo-Electron Microscopy Center for Structural Biology, Harvard Medical School, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Susanne von Gronau
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - J Wade Harper
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - John Hanna
- Department of Pathology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA.
| | - Brenda A Schulman
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
9
|
Zhou X, Xu R, Wu Y, Zhou L, Xiang T. The role of proteasomes in tumorigenesis. Genes Dis 2024; 11:101070. [PMID: 38523673 PMCID: PMC10958230 DOI: 10.1016/j.gendis.2023.06.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/10/2023] [Accepted: 06/27/2023] [Indexed: 03/26/2024] Open
Abstract
Protein homeostasis is the basis of normal life activities, and the proteasome family plays an extremely important function in this process. The proteasome 20S is a concentric circle structure with two α rings and two β rings overlapped. The proteasome 20S can perform both ATP-dependent and non-ATP-dependent ubiquitination proteasome degradation by binding to various subunits (such as 19S, 11S, and 200 PA), which is performed by its active subunit β1, β2, and β5. The proteasome can degrade misfolded, excess proteins to maintain homeostasis. At the same time, it can be utilized by tumors to degrade over-proliferate and unwanted proteins to support their growth. Proteasomes can affect the development of tumors from several aspects including tumor signaling pathways such as NF-κB and p53, cell cycle, immune regulation, and drug resistance. Proteasome-encoding genes have been found to be overexpressed in a variety of tumors, providing a potential novel target for cancer therapy. In addition, proteasome inhibitors such as bortezomib, carfilzomib, and ixazomib have been put into clinical application as the first-line treatment of multiple myeloma. More and more studies have shown that it also has different therapeutic effects in other tumors such as hepatocellular carcinoma, non-small cell lung cancer, glioblastoma, and neuroblastoma. However, proteasome inhibitors are not much effective due to their tolerance and singleness in other tumors. Therefore, further studies on their mechanisms of action and drug interactions are needed to investigate their therapeutic potential.
Collapse
Affiliation(s)
- Xiangyi Zhou
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Ruqing Xu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yue Wu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Li Zhou
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Tingxiu Xiang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
10
|
Rana PS, Ignatz-Hoover JJ, Kim BG, Malek E, Federov Y, Adams D, Chan T, Driscoll JJ. HDAC6 Inhibition Releases HR23B to Activate Proteasomes, Expand the Tumor Immunopeptidome and Amplify T-cell Antimyeloma Activity. CANCER RESEARCH COMMUNICATIONS 2024; 4:1517-1532. [PMID: 38747592 PMCID: PMC11188874 DOI: 10.1158/2767-9764.crc-23-0528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/14/2024] [Accepted: 05/09/2024] [Indexed: 06/19/2024]
Abstract
Proteasomes degrade intracellular proteins to generate antigenic peptides that are recognized by the adaptive immune system and promote anticancer immunity. However, tumors subvert the antigen presentation machinery to escape immunosurveillance. We hypothesized that proteasome activation could concomitantly increase antigen abundance and diversity in multiple myeloma cells. High-throughput screens revealed that histone deacetylase 6 (HDAC6) inhibitors activated proteasomes to unmask neoantigens and amplify the tumor-specific antigenic landscape. Treatment of patient CD138+ cells with HDAC6 inhibitors significantly promoted the antimyeloma activity of autologous CD8+ T cells. Pharmacologic blockade and genetic ablation of the HDAC6 ubiquitin-binding domain released HR23B, which shuttles ubiquitinylated cargo to proteasomes, while silencing HDAC6 or HR23B in multiple myeloma cells abolished the effect of HDAC6 inhibitors on proteasomes, antigen presentation, and T-cell cytotoxicity. Taken together, our results demonstrate the paradigm-shifting translational impact of proteasome activators to expand the myeloma immunopeptidome and have revealed novel, actionable antigenic targets for T cell-directed immunotherapy. SIGNIFICANCE The elimination of therapy-resistant tumor cells remains a major challenge in the treatment of multiple myeloma. Our study identifies and functionally validates agents that amplify MHC class I-presented antigens and pave the way for the development of proteasome activators as immune adjuvants to enhance immunotherapeutic responses in patients with multiple myeloma.
Collapse
Affiliation(s)
- Priyanka S. Rana
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - James J. Ignatz-Hoover
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Byung-Gyu Kim
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Ehsan Malek
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
- Adult Hematologic Malignancies and Stem Cell Transplant Section, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Yuriy Federov
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Small Molecule Drug Discovery Core, Case Western Reserve University, Cleveland, Ohio
| | - Drew Adams
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Small Molecule Drug Discovery Core, Case Western Reserve University, Cleveland, Ohio
| | - Timothy Chan
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Cleveland Clinic, Lerner Research Institute, Cleveland, Ohio
| | - James J. Driscoll
- Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio
- Adult Hematologic Malignancies and Stem Cell Transplant Section, Seidman Cancer Center, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| |
Collapse
|
11
|
Wen P, Sun Y, Jiang TX, Qiu XB. PA200-Mediated Proteasomal Protein Degradation and Regulation of Cellular Senescence. Int J Mol Sci 2024; 25:5637. [PMID: 38891826 PMCID: PMC11171664 DOI: 10.3390/ijms25115637] [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: 03/10/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 06/21/2024] Open
Abstract
Cellular senescence is closely related to DNA damage, proteasome inactivity, histone loss, epigenetic alterations, and tumorigenesis. The mammalian proteasome activator PA200 (also referred to as PSME4) or its yeast ortholog Blm10 promotes the acetylation-dependent degradation of the core histones during transcription, DNA repair, and spermatogenesis. According to recent studies, PA200 plays an important role in senescence, probably because of its role in promoting the degradation of the core histones. Loss of PA200 or Blm10 is a major cause of the decrease in proteasome activity during senescence. In this paper, recent research progress on the association of PA200 with cellular senescence is summarized, and the potential of PA200 to serve as a therapeutic target in age-related diseases is discussed.
Collapse
Affiliation(s)
- Pei Wen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; (P.W.); (Y.S.)
| | - Yan Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; (P.W.); (Y.S.)
| | - Tian-Xia Jiang
- Ministry of Education Key Laboratory of Cell Proliferation & Regulation Biology, College of Life Sciences, Beijing Normal University, 19 Xinjiekouwai Avenue, Beijing 100875, China
| | - Xiao-Bo Qiu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; (P.W.); (Y.S.)
- Ministry of Education Key Laboratory of Cell Proliferation & Regulation Biology, College of Life Sciences, Beijing Normal University, 19 Xinjiekouwai Avenue, Beijing 100875, China
| |
Collapse
|
12
|
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
|
13
|
Kandel R, Jung J, Neal S. Proteotoxic stress and the ubiquitin proteasome system. Semin Cell Dev Biol 2024; 156:107-120. [PMID: 37734998 PMCID: PMC10807858 DOI: 10.1016/j.semcdb.2023.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/01/2023] [Accepted: 08/20/2023] [Indexed: 09/23/2023]
Abstract
The ubiquitin proteasome system maintains protein homeostasis by regulating the breakdown of misfolded proteins, thereby preventing misfolded protein aggregates. The efficient elimination is vital for preventing damage to the cell by misfolded proteins, known as proteotoxic stress. Proteotoxic stress can lead to the collapse of protein homeostasis and can alter the function of the ubiquitin proteasome system. Conversely, impairment of the ubiquitin proteasome system can also cause proteotoxic stress and disrupt protein homeostasis. This review examines two impacts of proteotoxic stress, 1) disruptions to ubiquitin homeostasis (ubiquitin stress) and 2) disruptions to proteasome homeostasis (proteasome stress). Here, we provide a mechanistic description of the relationship between proteotoxic stress and the ubiquitin proteasome system. This relationship is illustrated by findings from several protein misfolding diseases, mainly neurodegenerative diseases, as well as from basic biology discoveries from yeast to mammals. In addition, we explore the importance of the ubiquitin proteasome system in endoplasmic reticulum quality control, and how proteotoxic stress at this organelle is alleviated. Finally, we highlight how cells utilize the ubiquitin proteasome system to adapt to proteotoxic stress and how the ubiquitin proteasome system can be genetically and pharmacologically manipulated to maintain protein homeostasis.
Collapse
Affiliation(s)
- Rachel Kandel
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California San Diego, La Jolla, CA 92093, United States
| | - Jasmine Jung
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California San Diego, La Jolla, CA 92093, United States
| | - Sonya Neal
- School of Biological Sciences, Department of Cell and Developmental Biology, University of California San Diego, La Jolla, CA 92093, United States; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
14
|
Morstein J, Amatuni A, Shuster A, Kuttenlochner W, Ko T, Abegg D, Groll M, Adibekian A, Renata H, Trauner DH. Optical Control of Proteasomal Protein Degradation with a Photoswitchable Lipopeptide. Angew Chem Int Ed Engl 2024; 63:e202314791. [PMID: 38109686 DOI: 10.1002/anie.202314791] [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: 10/02/2023] [Revised: 12/02/2023] [Accepted: 12/05/2023] [Indexed: 12/20/2023]
Abstract
Photolipids have emerged as attractive tools for the optical control of lipid functions. They often contain an azobenzene photoswitch that imparts a cis double-bond upon irradiation. Herein, we present the application of photoswitching to a lipidated natural product, the potent proteasome inhibitor cepafungin I. Several azobenzene-containing lipids were attached to the cyclopeptide core, yielding photoswitchable derivatives. Most notably, PhotoCep4 exhibited a 10-fold higher cellular potency in its light-induced cis-form, matching the potency of natural cepafungin I. The length of the photolipid tail and distal positioning of the azobenzene photoswitch with respect to the macrocycle is critical for this activity. In a proteome-wide experiment, light-triggered PhotoCep4 modulation showed high overlap with constitutively active cepafungin I. The mode of action was studied using crystallography and revealed an identical binding of the cyclopeptide in comparison to cepafungin I, suggesting that differences in their cellular activity originate from switching the tail structure. The photopharmacological approach described herein could be applicable to many other natural products as lipid conjugation is common and often necessary for potent activity. Such lipids are often introduced late in synthetic routes, enabling facile chemical modifications.
Collapse
Affiliation(s)
- Johannes Morstein
- Department of Cellular and Molecular Pharmacology and Howard Hughes Medical Institute, University of California, San Francisco, CA-94158, USA
- Department of Chemistry, New York University, New York, NY-10003, USA
| | - Alexander Amatuni
- Skaggs Doctoral Program in the Chemical and Biological Sciences, Scripps Research, La Jolla, CA-92037, USA
| | - Anton Shuster
- Skaggs Doctoral Program in the Chemical and Biological Sciences, Scripps Research, La Jolla, CA-92037, USA
| | - Wolfgang Kuttenlochner
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Center for Protein Assemblies, Chair of Biochemistry, Ernst-Otto-Fischer-Str. 8, 85748, Garching, Germany
| | - Tongil Ko
- Department of Chemistry, New York University, New York, NY-10003, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA-19104, USA
| | - Daniel Abegg
- Department of Chemistry, University of Illinois Chicago, Chicago, IL-60607, USA
| | - Michael Groll
- Technical University of Munich, TUM School of Natural Sciences, Department of Bioscience, Center for Protein Assemblies, Chair of Biochemistry, Ernst-Otto-Fischer-Str. 8, 85748, Garching, Germany
| | - Alexander Adibekian
- Department of Chemistry, University of Illinois Chicago, Chicago, IL-60607, USA
| | - Hans Renata
- Department of Chemistry, BioScience Research Collaborative, Rice University, Houston, TX-77005, USA
| | - Dirk H Trauner
- Department of Chemistry, New York University, New York, NY-10003, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, PA-19104, USA
| |
Collapse
|
15
|
Adolf F, Du J, Goodall EA, Walsh RM, Rawson S, von Gronau S, Harper JW, Hanna J, Schulman BA. Visualizing chaperone-mediated multistep assembly of the human 20S proteasome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.27.577538. [PMID: 38328185 PMCID: PMC10849659 DOI: 10.1101/2024.01.27.577538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Dedicated assembly factors orchestrate stepwise production of many molecular machines, including the 28-subunit proteasome core particle (CP) that mediates protein degradation. Here, we report cryo-EM reconstructions of seven recombinant human subcomplexes that visualize all five chaperones and the three active site propeptides across a wide swath of the assembly pathway. Comparison of these chaperone-bound intermediates and a matching mature CP reveals molecular mechanisms determining the order of successive subunit additions, and how proteasome subcomplexes and assembly factors structurally adapt upon progressive subunit incorporation to stabilize intermediates, facilitate the formation of subsequent intermediates, and ultimately rearrange to coordinate proteolytic activation with gated access to active sites. The structural findings reported here explain many previous biochemical and genetic observations. This work establishes a methodologic approach for structural analysis of multiprotein complex assembly intermediates, illuminates specific functions of assembly factors, and reveals conceptual principles underlying human proteasome biogenesis.
Collapse
Affiliation(s)
- Frank Adolf
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Jiale Du
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Ellen A. Goodall
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Richard M. Walsh
- Harvard Cryo-Electron Microscopy Center for Structural Biology, Harvard Medical School, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Shaun Rawson
- Harvard Cryo-Electron Microscopy Center for Structural Biology, Harvard Medical School, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Susanne von Gronau
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
| | - J. Wade Harper
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - John Hanna
- Department of Pathology, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Brenda A. Schulman
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| |
Collapse
|
16
|
Appasamy SD, Berrisford J, Gaborova R, Nair S, Anyango S, Grudinin S, Deshpande M, Armstrong D, Pidruchna I, Ellaway JIJ, Leines GD, Gupta D, Harrus D, Varadi M, Velankar S. Annotating Macromolecular Complexes in the Protein Data Bank: Improving the FAIRness of Structure Data. Sci Data 2023; 10:853. [PMID: 38040737 PMCID: PMC10692154 DOI: 10.1038/s41597-023-02778-9] [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: 05/15/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023] Open
Abstract
Macromolecular complexes are essential functional units in nearly all cellular processes, and their atomic-level understanding is critical for elucidating and modulating molecular mechanisms. The Protein Data Bank (PDB) serves as the global repository for experimentally determined structures of macromolecules. Structural data in the PDB offer valuable insights into the dynamics, conformation, and functional states of biological assemblies. However, the current annotation practices lack standardised naming conventions for assemblies in the PDB, complicating the identification of instances representing the same assembly. In this study, we introduce a method leveraging resources external to PDB, such as the Complex Portal, UniProt and Gene Ontology, to describe assemblies and contextualise them within their biological settings accurately. Employing the proposed approach, we assigned standard names to over 90% of unique assemblies in the PDB and provided persistent identifiers for each assembly. This standardisation of assembly data enhances the PDB, facilitating a deeper understanding of macromolecular complexes. Furthermore, the data standardisation improves the PDB's FAIR attributes, fostering more effective basic and translational research and scientific education.
Collapse
Affiliation(s)
- Sri Devan Appasamy
- Protein Data Bank in Europe, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK.
| | - John Berrisford
- Protein Data Bank in Europe, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Romana Gaborova
- CEITEC - Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Sreenath Nair
- Protein Data Bank in Europe, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Stephen Anyango
- Protein Data Bank in Europe, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Sergei Grudinin
- Univ. Grenoble Alpes, CNRS, Grenoble INP, LJK, 38000, Grenoble, France
| | - Mandar Deshpande
- Protein Data Bank in Europe, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - David Armstrong
- Protein Data Bank in Europe, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Ivanna Pidruchna
- Protein Data Bank in Europe, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Joseph I J Ellaway
- Protein Data Bank in Europe, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Grisell Díaz Leines
- Protein Data Bank in Europe, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Deepti Gupta
- Protein Data Bank in Europe, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Deborah Harrus
- Protein Data Bank in Europe, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Mihaly Varadi
- Protein Data Bank in Europe, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| | - Sameer Velankar
- Protein Data Bank in Europe, European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SD, UK
| |
Collapse
|
17
|
Zafeiropoulou K, Kalampounias G, Alexis S, Androutsopoulou T, Katsoris P, Symeonidis A. Lower-Risk Myelodysplastic Syndrome (MDS) Patients Exhibit Diminished Proteasome Proteolytic Activity and High Intracellular Reactive Oxygen Species (ROS) Levels. Cureus 2023; 15:e49843. [PMID: 38169896 PMCID: PMC10758539 DOI: 10.7759/cureus.49843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2023] [Indexed: 01/05/2024] Open
Abstract
Myelodysplastic syndromes (MDS) constitute a heterogeneous group of clonal hematopoietic stem cell disorders characterized by ineffective hematopoiesis and an elevated risk of transformation to acute myeloid leukemia (AML). Available disease-modifying treatment approaches are limited. The ineffectiveness of proteasome inhibitors (PIs) in MDS patients is currently investigated, although it is unclear whether they rapidly develop resistance to PIs or whether proteasome proteolytic activity (PPA) is constitutively lower in the hematopoietic cells of these patients, thus limiting treatment effectiveness. We investigated 20 patients with MDS, categorized according to the International Prognostic Scoring System (IPSS) into a lower- or a higher-risk group. Peripheral blood mononuclear cells, bone marrow mononuclear cells, and cluster of differentiation 34-positive (CD34+) cells were isolated and assessed for the chymotrypsin-like activity of the proteasome and β5 subunit accumulation. Additionally, intracellular reactive oxygen species (ROS) generation was screened. The lower-risk patient group (n=10) exhibited significantly lower proteasome activity (p<0.001) compared to both the higher-risk group (n=10) and healthy subjects (n=10). Furthermore, the lower-risk group had elevated oxidative stress levels (p<0.0001) and reduced β5 subunit expression (p=0.0286). Both parameters were shown to be associated with transfusion dependency, since transfusion-dependent patients (n=5 in each subgroup) had decreased proteasome activity and simultaneously exhibited higher ROS levels. Our results indicate that reduced β5 expression might potentially explain PIs' ineffectiveness in lower-risk MDS, elucidating the importance of the risk group in the selection of the proper treatment algorithm.
Collapse
Affiliation(s)
| | | | | | | | | | - Argiris Symeonidis
- School of Medicine, University of Patras, Patras, GRC
- Hematology Division, Department of Internal Medicine, University General Hospital of Patras, Patras, GRC
| |
Collapse
|
18
|
Park HM, Le L, Nguyen TT, Nam KH, Ordureau A, Lee JE, Nguyen TV. The CRL3 gigaxonin ubiquitin ligase-USP15 pathway governs the destruction of neurofilament proteins. Proc Natl Acad Sci U S A 2023; 120:e2306395120. [PMID: 37903270 PMCID: PMC10636361 DOI: 10.1073/pnas.2306395120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 09/20/2023] [Indexed: 11/01/2023] Open
Abstract
Giant axonal neuropathy (GAN) is caused by mutations in the GAN gene encoding for gigaxonin (GIG), which functions as an adaptor of the CUL3-RBX1-GIG (CRL3GIG) E3 ubiquitin ligase complex. The pathological hallmark of GAN is characterized by the accumulation of densely packed neurofilaments (NFs) in the axons. However, there are fundamental knowledge gaps in our understanding of the molecular mechanisms by which the ubiquitin-proteasome system controls the homeostasis of NF proteins. Recently, the deubiquitylating enzyme USP15 was reported to play a crucial role in regulating ubiquitylation and proteasomal degradation of CRL4CRBN substrate proteins. Here, we report that the CRL3GIG-USP15 pathway governs the destruction of NF proteins NEFL and INA. We identified a specific degron called NEFLL12 degron for CRL3GIG. Notably, mutations in the C-terminal Kelch domain of GIG, represented by L309R, R545C, and C570Y, disrupted the binding of GIG to NEFL and INA, leading to the accumulation of these NF proteins. This accounts for the loss-of-function mutations in GAN patients. In addition to regulating NFs, CRL3GIG also controls actin filaments by directly targeting actin-filament-binding regulatory proteins TPM1, TPM2, TAGLN, and CNN2 for proteasomal degradation. Thus, our findings broadly impact the field by providing fundamental mechanistic insights into regulating extremely long-lived NF proteins NEFL and INA by the CRL3GIG-USP15 pathway and offering previously unexplored therapeutic opportunities to treat GAN patients and other neurodegenerative diseases by explicitly targeting downstream substrates of CRL3GIG.
Collapse
Affiliation(s)
- Hyoung-Min Park
- Biometrology Group, Korea Research Institute of Standards and Science, Daejeon34113, Korea
| | - Ly Le
- Division of Quantum Simulation and Optimization, SandboxAQ, New York, NY10591
| | - Thao T. Nguyen
- Gehrke Proteomics Center, Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO65211
| | - Ki Hong Nam
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY10065
| | - Alban Ordureau
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY10065
| | - J. Eugene Lee
- Biometrology Group, Korea Research Institute of Standards and Science, Daejeon34113, Korea
| | - Thang Van Nguyen
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, MO65212
| |
Collapse
|
19
|
Terui H, Segawa Y, Asano Y. Targeting B cells for treatment of systemic sclerosis. Curr Opin Rheumatol 2023; 35:317-323. [PMID: 37540776 DOI: 10.1097/bor.0000000000000961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/06/2023]
Abstract
PURPOSE OF REVIEW The pathogenesis of systemic sclerosis (SSc) has been linked to dysfunctional B cells as demonstrated in previous research. This review aims to show the evidence and ongoing clinical trials of B cell-targeted therapy and overview the various aspects of B cell involvement in SSc. RECENT FINDINGS We provide an overview of the current understanding and therapeutic strategies targeting B cells in SSc patients. Several molecular targets of B cells have been identified for treating SSc, including CD20, CD19, B-cell activating factor (BAFF), and proteasome. SUMMARY Many clinical trials have demonstrated that B cells play a critical role in the pathogenesis of SSc and may be a potential therapeutic target to improve disease symptoms. Although large-scale clinical studies are needed, various B cell-targeted therapies have the potential to address the unmet needs of SSc patients.
Collapse
Affiliation(s)
- Hitoshi Terui
- Department of Dermatology, Tohoku University Graduate School of Medicine, Japan
| | | | | |
Collapse
|
20
|
Mostofian B, Martin HJ, Razavi A, Patel S, Allen B, Sherman W, Izaguirre JA. Targeted Protein Degradation: Advances, Challenges, and Prospects for Computational Methods. J Chem Inf Model 2023; 63:5408-5432. [PMID: 37602861 PMCID: PMC10498452 DOI: 10.1021/acs.jcim.3c00603] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Indexed: 08/22/2023]
Abstract
The therapeutic approach of targeted protein degradation (TPD) is gaining momentum due to its potentially superior effects compared with protein inhibition. Recent advancements in the biotech and pharmaceutical sectors have led to the development of compounds that are currently in human trials, with some showing promising clinical results. However, the use of computational tools in TPD is still limited, as it has distinct characteristics compared with traditional computational drug design methods. TPD involves creating a ternary structure (protein-degrader-ligase) responsible for the biological function, such as ubiquitination and subsequent proteasomal degradation, which depends on the spatial orientation of the protein of interest (POI) relative to E2-loaded ubiquitin. Modeling this structure necessitates a unique blend of tools initially developed for small molecules (e.g., docking) and biologics (e.g., protein-protein interaction modeling). Additionally, degrader molecules, particularly heterobifunctional degraders, are generally larger than conventional small molecule drugs, leading to challenges in determining drug-like properties like solubility and permeability. Furthermore, the catalytic nature of TPD makes occupancy-based modeling insufficient. TPD consists of multiple interconnected yet distinct steps, such as POI binding, E3 ligase binding, ternary structure interactions, ubiquitination, and degradation, along with traditional small molecule properties. A comprehensive set of tools is needed to address the dynamic nature of the induced proximity ternary complex and its implications for ubiquitination. In this Perspective, we discuss the current state of computational tools for TPD. We start by describing the series of steps involved in the degradation process and the experimental methods used to characterize them. Then, we delve into a detailed analysis of the computational tools employed in TPD. We also present an integrative approach that has proven successful for degrader design and its impact on project decisions. Finally, we examine the future prospects of computational methods in TPD and the areas with the greatest potential for impact.
Collapse
Affiliation(s)
- Barmak Mostofian
- OpenEye, Cadence Molecular Sciences, Boston, Massachusetts 02114 United States
| | - Holli-Joi Martin
- Laboratory
for Molecular Modeling, Division of Chemical Biology and Medicinal
Chemistry, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599 United States
| | - Asghar Razavi
- ENKO
Chem, Inc, Mystic, Connecticut 06355 United States
| | - Shivam Patel
- Psivant
Therapeutics, Boston, Massachusetts 02210 United States
| | - Bryce Allen
- Differentiated
Therapeutics, San Diego, California 92056 United States
| | - Woody Sherman
- Psivant
Therapeutics, Boston, Massachusetts 02210 United States
| | - Jesus A Izaguirre
- Differentiated
Therapeutics, San Diego, California 92056 United States
- Atommap
Corporation, New York, New York 10013 United States
| |
Collapse
|
21
|
Hsu HC, Wang J, Kjellgren A, Li H, DeMartino GN. Ηigh-resolution structure of mammalian PI31-20S proteasome complex reveals mechanism of proteasome inhibition. J Biol Chem 2023; 299:104862. [PMID: 37236357 PMCID: PMC10319324 DOI: 10.1016/j.jbc.2023.104862] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 05/08/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
Proteasome-catalyzed protein degradation mediates and regulates critical aspects of many cellular functions and is an important element of proteostasis in health and disease. Proteasome function is determined in part by the types of proteasome holoenzymes formed between the 20S core particle that catalyzes peptide bond hydrolysis and any of multiple regulatory proteins to which it binds. One of these regulators, PI31, was previously identified as an in vitro 20S proteasome inhibitor, but neither the molecular mechanism nor the possible physiologic significance of PI31-mediated proteasome inhibition has been clear. Here we report a high-resolution cryo-EM structure of the mammalian 20S proteasome in complex with PI31. The structure shows that two copies of the intrinsically disordered carboxyl terminus of PI31 are present in the central cavity of the closed-gate conformation of the proteasome and interact with proteasome catalytic sites in a manner that blocks proteolysis of substrates but resists their own degradation. The two inhibitory polypeptide chains appear to originate from PI31 monomers that enter the catalytic chamber from opposite ends of the 20S cylinder. We present evidence that PI31 can inhibit proteasome activity in mammalian cells and may serve regulatory functions for the control of cellular proteostasis.
Collapse
Affiliation(s)
- Hao-Chi Hsu
- Department of Structural Biology, Van Andel Institute, Grand Rapids, Michigan, USA
| | - Jason Wang
- Department of Physiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Abbey Kjellgren
- Department of Physiology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Huilin Li
- Department of Structural Biology, Van Andel Institute, Grand Rapids, Michigan, USA.
| | - George N DeMartino
- Department of Physiology, UT Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
22
|
Deshmukh FK, Ben-Nissan G, Olshina MA, Füzesi-Levi MG, Polkinghorn C, Arkind G, Leushkin Y, Fainer I, Fleishman SJ, Tawfik D, Sharon M. Allosteric regulation of the 20S proteasome by the Catalytic Core Regulators (CCRs) family. Nat Commun 2023; 14:3126. [PMID: 37253751 DOI: 10.1038/s41467-023-38404-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 04/26/2023] [Indexed: 06/01/2023] Open
Abstract
Controlled degradation of proteins is necessary for ensuring their abundance and sustaining a healthy and accurately functioning proteome. One of the degradation routes involves the uncapped 20S proteasome, which cleaves proteins with a partially unfolded region, including those that are damaged or contain intrinsically disordered regions. This degradation route is tightly controlled by a recently discovered family of proteins named Catalytic Core Regulators (CCRs). Here, we show that CCRs function through an allosteric mechanism, coupling the physical binding of the PSMB4 β-subunit with attenuation of the complex's three proteolytic activities. In addition, by dissecting the structural properties that are required for CCR-like function, we could recapitulate this activity using a designed protein that is half the size of natural CCRs. These data uncover an allosteric path that does not involve the proteasome's enzymatic subunits but rather propagates through the non-catalytic subunit PSMB4. This way of 20S proteasome-specific attenuation opens avenues for decoupling the 20S and 26S proteasome degradation pathways as well as for developing selective 20S proteasome inhibitors.
Collapse
Affiliation(s)
- Fanindra Kumar Deshmukh
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Gili Ben-Nissan
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Maya A Olshina
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Maria G Füzesi-Levi
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Caley Polkinghorn
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Galina Arkind
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Yegor Leushkin
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Irit Fainer
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Sarel J Fleishman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Dan Tawfik
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Michal Sharon
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, 7610001, Israel.
| |
Collapse
|
23
|
Gong C, Bonfili L, Zheng Y, Cecarini V, Cuccioloni M, Angeletti M, Dematteis G, Tapella L, Genazzani AA, Lim D, Eleuteri AM. Immortalized Alzheimer's Disease Astrocytes: Characterization of Their Proteolytic Systems. Mol Neurobiol 2023; 60:2787-2800. [PMID: 36729287 PMCID: PMC10039838 DOI: 10.1007/s12035-023-03231-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/12/2023] [Indexed: 02/03/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegeneration with dysfunctions in both the ubiquitin-proteasome system (UPS) and autophagy. Astroglia participation in AD is an attractive topic of research, but molecular patterns are partially defined and available in vitro models have technical limitations. Immortalized astrocytes from the hippocampus of 3xTg-AD and wild-type mice (3Tg-iAstro and WT-iAstro, respectively) have been obtained as an attempt to overcome primary cell line limitations and this study aims at characterizing their proteolytic systems, focusing on UPS and autophagy. Both 26S and 20S proteasomal activities were downregulated in 3Tg-iAstro, in which a shift in catalytic subunits from constitutive 20S proteasome to immunoproteasome occurred, with consequences on immune functions. In fact, immunoproteasome is the specific complex in charge of clearing damaged proteins under inflammatory conditions. Parallelly, augmented expression and activity of the lysosomal cathepsin B, enhanced levels of lysosomal-associated membrane protein 1, beclin1, and LC3-II, together with an increased uptake of monodansylcadaverine in autophagic vacuoles, suggested autophagy activation in 3Tg-iAstro. The two proteolytic pathways were linked by p62 that accumulated in 3Tg-iAstro due to both increased synthesis and decreased degradation in the UPS defective astrocytes. Treatment with 4-phenylbutyric acid, a neuroprotective small chemical chaperone, partially restored proteasome and autophagy-mediated proteolysis in 3Tg-iAstro. Our data shed light on the impaired proteostasis in 3Tg-iAstro with proteasome inhibition and autophagic compensatory activation, providing additional validation of this AD in vitro model, and propose a new mechanism of action of 4-phenylbutyric acid in neurodegenerative disorders.
Collapse
Affiliation(s)
- Chunmei Gong
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, MC, Italy
| | - Laura Bonfili
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, MC, Italy.
| | - Yadong Zheng
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, MC, Italy
| | - Valentina Cecarini
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, MC, Italy
| | - Massimiliano Cuccioloni
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, MC, Italy
| | - Mauro Angeletti
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, MC, Italy
| | - Giulia Dematteis
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Via Bovio 6, 28100, Novara, Italy
| | - Laura Tapella
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Via Bovio 6, 28100, Novara, Italy
| | - Armando A Genazzani
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Via Bovio 6, 28100, Novara, Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, Via Bovio 6, 28100, Novara, Italy.
| | - Anna Maria Eleuteri
- School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, MC, Italy.
| |
Collapse
|
24
|
Hsu HC, Wang J, Kjellgren A, Li H, DeMartino GN. High-resolution structure of mammalian PI31â€"20S proteasome complex reveals mechanism of proteasome inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.535455. [PMID: 37066326 PMCID: PMC10103979 DOI: 10.1101/2023.04.03.535455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Proteasome-catalyzed protein degradation mediates and regulates critical aspects of many cellular functions and is an important element of proteostasis in health and disease. Proteasome function is determined in part by the types of proteasome holoenzymes formed between the 20S core particle that catalyzes peptide bond hydrolysis and any of multiple regulatory proteins to which it binds. One of these regulators, PI31, was previously identified as an in vitro 20S proteasome inhibitor, but neither the molecular mechanism nor the possible physiologic significance of PI31-mediated proteasome inhibition has been clear. Here we report a high- resolution cryo-EM structure of the mammalian 20S proteasome in complex with PI31. The structure shows that two copies of the intrinsically-disordered carboxyl-terminus of PI31 are present in the central cavity of the closed-gate conformation of the proteasome and interact with proteasome catalytic sites in a manner that blocks proteolysis of substrates but resists their own degradation. The two inhibitory polypeptide chains appear to originate from PI31 monomers that enter the catalytic chamber from opposite ends of the 20S cylinder. We present evidence that PI31 can inhibit proteasome activity in mammalian cells and may serve regulatory functions for the control of cellular proteostasis.
Collapse
|
25
|
Sahu I, Bajorek M, Tan X, Srividya M, Krutauz D, Reis N, Osmulski PA, Gaczynska ME, Glickman MH. A Role for the Proteasome Alpha2 Subunit N-Tail in Substrate Processing. Biomolecules 2023; 13:480. [PMID: 36979414 PMCID: PMC10046698 DOI: 10.3390/biom13030480] [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: 01/19/2023] [Revised: 02/23/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
The proteolytic active sites of the 26S proteasome are sequestered within the catalytic chamber of its 20S core particle (CP). Access to this chamber is through a narrow channel defined by the seven outer α subunits. In the resting state, the N-termini of neighboring α subunits form a gate blocking access to the channel. The attachment of the activators or regulatory particles rearranges the blocking α subunit N-termini facilitating the entry of substrates. By truncating or mutating each of the participating α N-termini, we report that whereas only a few N-termini are important for maintaining the closed gate, all seven N-termini participate in the open gate. Specifically, the open state is stabilized by a hydrogen bond between an invariant tyrosine (Y) in each subunit with a conserved aspartate (D) in its counterclockwise neighbor. The lone exception is the α1-α2 pair leaving a gap in the ring circumference. The third residue (X) of this YD(X) motif aligns with the open channel. Phenylalanine at this position in the α2 subunit comes in direct contact with the translocating substrate. Consequently, deletion of the α2 N-terminal tail attenuates proteolysis despite the appearance of an open gate state. In summary, the interlacing N-terminal YD(X) motifs regulate both the gating and translocation of the substrate.
Collapse
Affiliation(s)
- Indrajit Sahu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Monika Bajorek
- Faculty of Biology, Technion—Israel Institute of Technology, Haifa 3525433, Israel
| | - Xiaolin Tan
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Madabhushi Srividya
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Daria Krutauz
- Faculty of Biology, Technion—Israel Institute of Technology, Haifa 3525433, Israel
| | - Noa Reis
- Faculty of Biology, Technion—Israel Institute of Technology, Haifa 3525433, Israel
| | - Pawel A. Osmulski
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Maria E. Gaczynska
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Michael H. Glickman
- Faculty of Biology, Technion—Israel Institute of Technology, Haifa 3525433, Israel
| |
Collapse
|
26
|
Ang’ang’o LM, Herren JK, Tastan Bishop Ö. Structural and Functional Annotation of Hypothetical Proteins from the Microsporidia Species Vittaforma corneae ATCC 50505 Using in silico Approaches. Int J Mol Sci 2023; 24:3507. [PMID: 36834914 PMCID: PMC9960886 DOI: 10.3390/ijms24043507] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/25/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Microsporidia are spore-forming eukaryotes that are related to fungi but have unique traits that set them apart. They have compact genomes as a result of evolutionary gene loss associated with their complete dependency on hosts for survival. Despite having a relatively small number of genes, a disproportionately high percentage of the genes in microsporidia genomes code for proteins whose functions remain unknown (hypothetical proteins-HPs). Computational annotation of HPs has become a more efficient and cost-effective alternative to experimental investigation. This research developed a robust bioinformatics annotation pipeline of HPs from Vittaforma corneae, a clinically important microsporidian that causes ocular infections in immunocompromised individuals. Here, we describe various steps to retrieve sequences and homologs and to carry out physicochemical characterization, protein family classification, identification of motifs and domains, protein-protein interaction network analysis, and homology modelling using a variety of online resources. Classification of protein families produced consistent findings across platforms, demonstrating the accuracy of annotation utilizing in silico methods. A total of 162 out of 2034 HPs were fully annotated, with the bulk of them categorized as binding proteins, enzymes, or regulatory proteins. The protein functions of several HPs from Vittaforma corneae were accurately inferred. This improved our understanding of microsporidian HPs despite challenges related to the obligate nature of microsporidia, the absence of fully characterized genes, and the lack of homologous genes in other systems.
Collapse
Affiliation(s)
- Lilian Mbaisi Ang’ang’o
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry and Microbiology, Rhodes University, Makhanda 6140, South Africa
| | - Jeremy Keith Herren
- International Centre of Insect Physiology and Ecology (icipe), Nairobi P.O. Box 30772-00100, Kenya
| | - Özlem Tastan Bishop
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry and Microbiology, Rhodes University, Makhanda 6140, South Africa
| |
Collapse
|
27
|
Warnock JL, Jobin GW, Kumar S, Tomko RJ. Assembly chaperone Nas6 selectively destabilizes 26S proteasomes with defective regulatory particle-core particle interfaces. J Biol Chem 2023; 299:102894. [PMID: 36634850 PMCID: PMC9943895 DOI: 10.1016/j.jbc.2023.102894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 01/11/2023] Open
Abstract
The 26S proteasome is a 66-subunit-chambered protease present in all eukaryotes that maintains organismal health by degrading unneeded or defective proteins. Defects in proteasome function or assembly are known to contribute to the development of various cancers, neurodegeneration, and diabetes. During proteasome biogenesis, a family of evolutionarily conserved chaperones assembles a hexameric ring of AAA+ family ATPase subunits contained within the proteasomal regulatory particle (RP) and guide their docking onto the surface of the proteolytic core particle (CP). This RP-CP interaction couples the substrate capture and unfolding process to proteolysis. We previously reported a mutation in the proteasome that promoted dissociation of the RP and CP by one of these chaperones, Nas6. However, the nature of the signal for Nas6-dependent proteasome disassembly and the generality of this postassembly proteasome quality control function for Nas6 remain unknown. Here, we use structure-guided mutagenesis and in vitro proteasome disassembly assays to demonstrate that Nas6 more broadly destabilizes 26S proteasomes with a defective RP-CP interface. We show that Nas6 can promote dissociation of mature proteasomes into RP and CP in cells harboring defects on either side of the RP-CP interface. This function is unique to Nas6 and independent from other known RP assembly chaperones. Further biochemical experiments suggest that Nas6 may exploit a weakened RP-CP interface to dissociate the RP from the CP. We propose that this postassembly role of Nas6 may fulfill a quality control function in cells by promoting the recycling of functional subcomplexes contained within defective proteasomes.
Collapse
|
28
|
Bolik-Coulon N, Sever AIM, Harkness RW, Aramini JM, Toyama Y, Hansen DF, Kay LE. Less is more: A simple methyl-TROSY based pulse scheme offers improved sensitivity in applications to high molecular weight complexes. JOURNAL OF MAGNETIC RESONANCE (SAN DIEGO, CALIF. : 1997) 2023; 346:107326. [PMID: 36508761 DOI: 10.1016/j.jmr.2022.107326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 06/17/2023]
Abstract
The HMQC pulse sequence and variants thereof have been exploited in studies of high molecular weight protein complexes, taking advantage of the fact that fast and slow relaxing magnetization components are sequestered along two distinct magnetization transfer pathways. Despite the simplicity of the HMQC scheme an even shorter version can be designed, based on elimination of the terminal refocusing period, as a further means of increasing signal. Here we present such an experiment, and show that significant sensitivity gains, in some cases by factors of two or more, are realized in studies of proteins varying in molecular masses from 100 kDa to 1 MDa.
Collapse
Affiliation(s)
- Nicolas Bolik-Coulon
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | | | - Robert W Harkness
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - James M Aramini
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yuki Toyama
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - D Flemming Hansen
- Department of Structural and Molecular Biology, Division of Biosciences, University College London, London WC1E 6BT, United Kingdom.
| | - Lewis E Kay
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada; Program in Molecular Medicine, Hospital for Sick Children Research Institute, Toronto, ON M5G 0A4, Canada.
| |
Collapse
|
29
|
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
|
30
|
Amela-Cortes M, Wilmet M, Le Person S, Khlifi S, Lebastard C, Molard Y, Cordier S. From Solid-State Cluster Compounds to Functional PMMA-Based Composites with UV and NIR Blocking Properties, and Tuned Hues. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 13:144. [PMID: 36616054 PMCID: PMC9824331 DOI: 10.3390/nano13010144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 06/17/2023]
Abstract
New nanocomposite materials with UV-NIR blocking properties and hues ranging from green to brown were prepared by integrating inorganic tantalum octahedral cluster building blocks prepared via solid-state chemistry in a PMMA matrix. After the synthesis by the solid-state chemical reaction of the K4[{Ta6Bri12}Bra6] ternary halide, built-up from [{Ta6Bri12}Bra6]4- anionic building blocks, and potassium cations, the potassium cations were replaced by functional organic cations (Kat+) bearing a methacrylate function. The resulting intermediate, (Kat)2[{Ta6Bri12}Bra6], was then incorporated homogeneously by copolymerization with MMA into transparent PMMA matrices to form a brown transparent hybrid composite Ta@PMMAbrown. The color of the composites was tuned by controlling the charge and consequently the oxidation state of the cluster building block. Ta@PMMAgreen was obtained through the two-electron reduction of the [{Ta6Bri12}Bra6]2- building blocks from Ta@PMMAbrown in solution. Indeed, the control of the oxidation state of the Ta6 cluster inorganic building blocks occurred inside the copolymer, which not only allowed the tuning of the optical properties of the composite in the visible region but also allowed the tuning of its UV and NIR blocking properties.
Collapse
Affiliation(s)
| | - Maxence Wilmet
- CNRS-Saint Gobain-NIMS, IRL3629, Laboratory for Innovative Key Materials ans Structures (LINK), National Institute for Materials Science (NIMS), Tsukuba 305-0044, Japan
| | | | - Soumaya Khlifi
- Univ. Rennes, CNRS, ISCR, UMR6226, F-35000 Rennes, France
| | - Clément Lebastard
- Univ. Rennes, CNRS, ISCR, UMR6226, F-35000 Rennes, France
- CNRS-Saint Gobain-NIMS, IRL3629, Laboratory for Innovative Key Materials ans Structures (LINK), National Institute for Materials Science (NIMS), Tsukuba 305-0044, Japan
| | - Yann Molard
- Univ. Rennes, CNRS, ISCR, UMR6226, F-35000 Rennes, France
| | | |
Collapse
|
31
|
The Emerging Roles of Extracellular Chaperones in Complement Regulation. Cells 2022; 11:cells11233907. [PMID: 36497163 PMCID: PMC9738919 DOI: 10.3390/cells11233907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/01/2022] [Accepted: 12/01/2022] [Indexed: 12/09/2022] Open
Abstract
The immune system is essential to protect organisms from internal and external threats. The rapidly acting, non-specific innate immune system includes complement, which initiates an inflammatory cascade and can form pores in the membranes of target cells to induce cell lysis. Regulation of protein homeostasis (proteostasis) is essential for normal cellular and organismal function, and has been implicated in processes controlling immunity and infection. Chaperones are key players in maintaining proteostasis in both the intra- and extracellular environments. Whilst intracellular proteostasis is well-characterised, the role of constitutively secreted extracellular chaperones (ECs) is less well understood. ECs may interact with invading pathogens, and elements of the subsequent immune response, including the complement pathway. Both ECs and complement can influence the progression of neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease and amyotrophic lateral sclerosis, as well as other diseases including kidney diseases and diabetes. This review will examine known and recently discovered ECs, and their roles in immunity, with a specific focus on the complement pathway.
Collapse
|
32
|
Walsh CT. Covalent Catalytic Strategies for Enzymes That Modify RNA Molecules on their Tripartite Building Blocks. ACS Chem Biol 2022; 17:2686-2703. [PMID: 36103129 DOI: 10.1021/acschembio.2c00584] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The tripartite structures of the four 5'-nucleotide monophosphate (NMP) building blocks in all RNAs enable enzyme-catalyzed chemical modifications to three types of sites: the heterocyclic bases via N- and C-methylations and other alkylations, conversion of the N-glycoside linkages of the uridine moiety to the C-C glycoside link in pseudouridines, and the phosphodiester-mediated processes of 5'-capping, splicing, and 3'-tailing of premRNAs. We examine known cases for enzymatic covalent catalytic strategies that entail transient formation and breakdown of covalent enzyme-RNA adducts in each catalytic cycle. One case involves generation of the required carbon nucleophile during C5 methylation of cytosine residues in RNAs. A second examines the mechanism proposed for pseudouridine synthases and for replacement of a guanine residue in tRNAs by queuosine. The third category involves phosphoric anhydride and phosphodiester chemistry by which viral RNAs encode enzymes for making their own mRNA 5'-caps. This strategy includes the recent finding that the SARS-CoV2 proteins assemble a canonical 5',5'-GTP cap on their 28 900 nucleotide genomic RNA to enable its translation as an mRNA by host translational machinery by way of a covalent RNA-viral enzyme intermediate.
Collapse
Affiliation(s)
- Christopher T Walsh
- ChEM-H Institute, Stanford University, Palo Alto, California 94305, United States
| |
Collapse
|
33
|
Hernández JS, Shamshurin M, Puche M, Sokolov MN, Feliz M. Nanostructured Hybrids Based on Tantalum Bromide Octahedral Clusters and Graphene Oxide for Photocatalytic Hydrogen Evolution. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:3647. [PMID: 36296837 PMCID: PMC9611948 DOI: 10.3390/nano12203647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 06/16/2023]
Abstract
The generation of hydrogen (H2) using sunlight has become an essential energy alternative for decarbonization. The need for functional nanohybrid materials based on photo- and electroactive materials and accessible raw materials is high in the field of solar fuels. To reach this goal, single-step synthesis of {Ta6Bri12}@GO (GO = graphene oxide) nanohybrids was developed by immobilization of [{Ta6Bri12}Bra2(H2O)a4]·4H2O (i = inner and a = apical positions of the Ta6 octahedron) on GO nanosheets by taking the advantage of the easy ligand exchange of the apical cluster ligands with the oxygen functionalities of GO. The nanohybrids were characterized by spectroscopic, analytical, and morphological techniques. The hybrid formation enhances the yield of photocatalytic H2 from water with respect to their precursors and this is without the presence of precious metals. This enhancement is attributed to the optimal cluster loading onto the GO support and the crucial role of GO in the electron transfer from Ta6 clusters into GO sheets, thus suppressing the charge recombination. In view of the simplicity and versatility of the designed photocatalytic system, octahedral tantalum clusters are promising candidates to develop new and environmentally friendly photocatalysts for H2 evolution.
Collapse
Affiliation(s)
- Jhon Sebastián Hernández
- Instituto de Tecnología Química, Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas (UPV-CSIC), Avd. de los Naranjos s/n, 46022 Valencia, Spain
| | - Maxim Shamshurin
- Nikolaev Institute of Inorganic Chemistry SB RAS, 3 Akad. Lavrentiev Ave., 630090 Novosibirsk, Russia
| | - Marta Puche
- Instituto de Tecnología Química, Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas (UPV-CSIC), Avd. de los Naranjos s/n, 46022 Valencia, Spain
| | - Maxim N. Sokolov
- Nikolaev Institute of Inorganic Chemistry SB RAS, 3 Akad. Lavrentiev Ave., 630090 Novosibirsk, Russia
| | - Marta Feliz
- Instituto de Tecnología Química, Universitat Politècnica de València-Consejo Superior de Investigaciones Científicas (UPV-CSIC), Avd. de los Naranjos s/n, 46022 Valencia, Spain
| |
Collapse
|
34
|
Nguyen NTK, Lebastard C, Wilmet M, Dumait N, Renaud A, Cordier S, Ohashi N, Uchikoshi T, Grasset F. A review on functional nanoarchitectonics nanocomposites based on octahedral metal atom clusters (Nb 6, Mo 6, Ta 6, W 6, Re 6): inorganic 0D and 2D powders and films. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2022; 23:547-578. [PMID: 36212682 PMCID: PMC9542349 DOI: 10.1080/14686996.2022.2119101] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/10/2022] [Accepted: 08/24/2022] [Indexed: 05/29/2023]
Abstract
This review is dedicated to various functional nanoarchitectonic nanocomposites based on molecular octahedral metal atom clusters (Nb6, Mo6, Ta6, W6, Re6). Powder and film nanocomposites with two-dimensional, one-dimensional and zero-dimensional morphologies are presented, as well as film matrices from organic polymers to inorganic layered oxides. The high potential and synergetic effects of these nanocomposites for biotechnology applications, photovoltaic, solar control, catalytic, photonic and sensor applications are demonstrated. This review also provides a basic level of understanding how nanocomposites are characterized and processed using different techniques and methods. The main objective of this review would be to provide guiding significance for the design of new high-performance nanocomposites based on transition metal atom clusters.
Collapse
Affiliation(s)
- Ngan T. K. Nguyen
- CNRS-Saint Gobain-NIMS, IRL3629, Laboratory for Innovative Key Materials and Structures (LINK), National Institute for Materials Science (NIMS), Tsukuba, Japan
- International Center for Young Scientists, ICYS-Sengen, Global Networking Division, NIMS, Tsukuba, Japan
| | - Clément Lebastard
- CNRS-Saint Gobain-NIMS, IRL3629, Laboratory for Innovative Key Materials and Structures (LINK), National Institute for Materials Science (NIMS), Tsukuba, Japan
- Université Rennes, CNRS, ISCR, UMR6226, Rennes, France
| | - Maxence Wilmet
- CNRS-Saint Gobain-NIMS, IRL3629, Laboratory for Innovative Key Materials and Structures (LINK), National Institute for Materials Science (NIMS), Tsukuba, Japan
- Université Rennes, CNRS, ISCR, UMR6226, Rennes, France
- Saint Gobain Research Paris, Aubervilliers, France
| | - Noée Dumait
- Université Rennes, CNRS, ISCR, UMR6226, Rennes, France
| | - Adèle Renaud
- Université Rennes, CNRS, ISCR, UMR6226, Rennes, France
| | | | - Naoki Ohashi
- CNRS-Saint Gobain-NIMS, IRL3629, Laboratory for Innovative Key Materials and Structures (LINK), National Institute for Materials Science (NIMS), Tsukuba, Japan
- Research Center for Functional Materials, NIMS, Tsukuba, Japan
| | - Tetsuo Uchikoshi
- CNRS-Saint Gobain-NIMS, IRL3629, Laboratory for Innovative Key Materials and Structures (LINK), National Institute for Materials Science (NIMS), Tsukuba, Japan
- Research Center for Functional Materials, NIMS, Tsukuba, Japan
| | - Fabien Grasset
- CNRS-Saint Gobain-NIMS, IRL3629, Laboratory for Innovative Key Materials and Structures (LINK), National Institute for Materials Science (NIMS), Tsukuba, Japan
- Université Rennes, CNRS, ISCR, UMR6226, Rennes, France
| |
Collapse
|
35
|
Koester DC, Marx VM, Williams S, Jiricek J, Dauphinais M, René O, Miller SL, Zhang L, Patra D, Chen YL, Cheung H, Gable J, Lakshminarayana SB, Osborne C, Galarneau JR, Kulkarni U, Richmond W, Bretz A, Xiao L, Supek F, Wiesmann C, Honnappa S, Be C, Mäser P, Kaiser M, Ritchie R, Barrett MP, Diagana TT, Sarko C, Rao SPS. Discovery of Novel Quinoline-Based Proteasome Inhibitors for Human African Trypanosomiasis (HAT). J Med Chem 2022; 65:11776-11787. [PMID: 35993839 PMCID: PMC9469205 DOI: 10.1021/acs.jmedchem.2c00791] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Human African Trypanosomiasis (HAT) is a vector-borne disease caused by kinetoplastid parasites of the Trypanosoma genus. The disease proceeds in two stages, with a hemolymphatic blood stage and a meningo-encephalic brain stage. In the latter stage, the parasite causes irreversible damage to the brain leading to sleep cycle disruption and is fatal if untreated. An orally bioavailable treatment is highly desirable. In this study, we present a brain-penetrant, parasite-selective 20S proteasome inhibitor that was rapidly optimized from an HTS singleton hit to drug candidate compound 7 that showed cure in a stage II mouse efficacy model. Here, we describe hit expansion and lead optimization campaign guided by cryo-electron microscopy and an in silico model to predict the brain-to-plasma partition coefficient Kp as an important parameter to prioritize compounds for synthesis. The model combined with in vitro and in vivo experiments allowed us to advance compounds with favorable unbound brain-to-plasma ratios (Kp,uu) to cure a CNS disease such as HAT.
Collapse
Affiliation(s)
- Dennis C. Koester
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Vanessa M. Marx
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Sarah Williams
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Jan Jiricek
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Maxime Dauphinais
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Olivier René
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Sarah L. Miller
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Lei Zhang
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Debjani Patra
- Novartis
Institutes for Tropical Diseases, Emeryville, California 94608, United States
| | - Yen-Liang Chen
- Lead
Discovery, Novartis Institutes for Tropical
Diseases, Emeryville, California 94608, United States
| | - Harry Cheung
- Lead
Discovery, Novartis Institutes for Tropical
Diseases, Emeryville, California 94608, United States
| | - Jonathan Gable
- Lead
Discovery, Novartis Institutes for Tropical
Diseases, Emeryville, California 94608, United States
| | - Suresh B. Lakshminarayana
- Pharmacokinetic
Sciences, Novartis Institutes for Tropical
Diseases, Emeryville, California 94608, United States
| | - Colin Osborne
- Pharmacokinetic
Sciences, Pharmacology and Comparative Medicine, Novartis Institutes for Tropical Diseases, Emeryville, California 94608, United States
| | - Jean-Rene Galarneau
- Preclinical
Safety, Novartis Institutes for Biomedical
Research, Cambridge, Massachusetts 02139, United States
| | - Upendra Kulkarni
- Chemical
and Pharmaceutical Profiling, Novartis Institutes
for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Wendy Richmond
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, San Diego, California 92121, United States
| | - Angela Bretz
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, San Diego, California 92121, United States
| | - Linda Xiao
- Pharmacology, Novartis Institutes for Tropical Diseases, Emeryville, California 94608, United States
| | - Frantisek Supek
- Novartis
Institutes for Biomedical Research, San Diego, California 92121, United States
| | | | - Srinivas Honnappa
- Novartis
Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Celine Be
- Novartis
Institutes for Biomedical Research, 4056 Basel, Switzerland
| | - Pascal Mäser
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- University
of Basel, CH 4000 Basel, Switzerland
| | - Marcel Kaiser
- Swiss Tropical and Public Health Institute, Kreuzstrasse 2, 4123 Allschwil, Switzerland
- University
of Basel, CH 4000 Basel, Switzerland
| | - Ryan Ritchie
- University of Glasgow, University Place, Glasgow G12 8TA, U.K
| | | | - Thierry T. Diagana
- Novartis
Institutes for Tropical Diseases, Emeryville, California 94608, United States
| | - Christopher Sarko
- Global
Discovery Chemistry, Novartis Institutes
for Biomedical Research, Emeryville, California 94608, United States
| | - Srinivasa P. S. Rao
- Novartis
Institutes for Tropical Diseases, Emeryville, California 94608, United States
| |
Collapse
|
36
|
PKR Protects the Major Catalytic Subunit of PKA Cpk1 from FgBlm10-Mediated Proteasome Degradation in Fusarium graminearum. Int J Mol Sci 2022; 23:ijms231810208. [PMID: 36142119 PMCID: PMC9499325 DOI: 10.3390/ijms231810208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
For optimal proteolytic function, the proteasome core (CP or 20S) must associate with activators. The cAMP-PKA pathway is reported to affect the activity of the proteasome in humans. However, the relationship between the proteasome and PKA is not well characterized. Our results showed that the major catalytic subunit Cpk1 was degraded without the protection of Pkr. Eleven (out of 67) pkr suppressors had FgBlm10 C-terminal truncation, one suppressor had an amino acid change mutation in the PRE6 ortholog (FGRRES_07282), and one in the PRE5 ortholog (FGRRES_05222). These mutations rescued the defects in growth and conidial morphology, Cpk1 stability, and PKA activities in the pkr mutant. The interaction of FgBlm10 with FgPre5 and FgPre6 were detected by co-immunoprecipitation, and the essential elements for their interaction were characterized, including the FgBlm10 C-terminus, amino acid D82 of FgPre6 and K62 of FgPre5. Additional FgBlm10-interacting proteins were identified in the wild type and pkr mutant, suggesting that PKA regulates the preference of FgBlm10-mediated proteasome assembly. In addition, PKA indirectly affected the phosphorylation of FgBlm10, and its localization in the nucleus. The truncation of the FgBlm10 C terminus also enhanced nuclear import and bleomycin resistance, suggesting its role in proteasome assembly at DNA damage sites. Collectively, our data demonstrated that regulation between PKA and proteasome degradation is critical for the vegetative growth of F. graminearum.
Collapse
|
37
|
Ferrari V, Stroobant V, Abi Habib J, Naulaerts S, Van den Eynde BJ, Vigneron N. New Insights into the Mechanisms of Proteasome-Mediated Peptide Splicing Learned from Comparing Splicing Efficiency by Different Proteasome Subtypes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:2817-2828. [PMID: 35688464 DOI: 10.4049/jimmunol.2101198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 04/03/2022] [Indexed: 06/15/2023]
Abstract
By tying peptide fragments originally distant in parental proteins, the proteasome can generate spliced peptides that are recognized by CTL. This occurs by transpeptidation involving a peptide-acyl-enzyme intermediate and another peptide fragment present in the catalytic chamber. Four main subtypes of proteasomes exist: the standard proteasome (SP), the immunoproteasome, and intermediate proteasomes β1-β2-β5i (single intermediate proteasome) and β1i-β2-β5i (double intermediate proteasome). In this study, we use a tandem mass tag-quantification approach to study the production of six spliced human antigenic peptides by the four proteasome subtypes. Peptides fibroblast growth factor-5172-176/217-220, tyrosinase368-373/336-340, and gp10040-42/47-52 are better produced by the SP than the other proteasome subtypes. The peptides SP110296-301/286-289, gp100195-202/191or192, and gp10047-52/40-42 are better produced by the immunoproteasome and double intermediate proteasome. The current model of proteasome-catalyzed peptide splicing suggests that the production of a spliced peptide depends on the abundance of the peptide splicing partners. Surprisingly, we found that despite the fact that reciprocal peptides RTK_QLYPEW (gp10040-42/47-52) and QLYPEW_RTK (gp10047-52/40-42) are composed of identical splicing partners, their production varies differently according to the proteasome subtype. These differences were maintained after in vitro digestions involving identical amounts of the splicing fragments. Our results indicate that the amount of splicing partner is not the only factor driving peptide splicing and suggest that peptide splicing efficiency also relies on other factors, such as the affinity of the C-terminal splice reactant for the primed binding site of the catalytic subunit.
Collapse
Affiliation(s)
- Violette Ferrari
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Lifesciences and Biotechnology, Brussels, Belgium; and
| | - Vincent Stroobant
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Lifesciences and Biotechnology, Brussels, Belgium; and
| | - Joanna Abi Habib
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Lifesciences and Biotechnology, Brussels, Belgium; and
| | - Stefan Naulaerts
- Ludwig Institute for Cancer Research, Brussels, Belgium
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Lifesciences and Biotechnology, Brussels, Belgium; and
| | - Benoit J Van den Eynde
- Ludwig Institute for Cancer Research, Brussels, Belgium;
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Lifesciences and Biotechnology, Brussels, Belgium; and
- Nuffield Department of Medicine, Ludwig Institute for Cancer Research, University of Oxford, Oxford, United Kingdom
| | - Nathalie Vigneron
- Ludwig Institute for Cancer Research, Brussels, Belgium;
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Lifesciences and Biotechnology, Brussels, Belgium; and
| |
Collapse
|
38
|
Lebastard C, Wilmet M, Cordier S, Comby-Zerbino C, MacAleese L, Dugourd P, Hara T, Ohashi N, Uchikoshi T, Grasset F. Controlling the Deposition Process of Nanoarchitectonic Nanocomposites Based on {Nb 6-xTa xX i12} n+ Octahedral Cluster-Based Building Blocks (X i = Cl, Br; 0 ≤ x ≤ 6, n = 2, 3, 4) for UV-NIR Blockers Coating Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2052. [PMID: 35745391 PMCID: PMC9227475 DOI: 10.3390/nano12122052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/30/2022] [Accepted: 06/03/2022] [Indexed: 12/20/2022]
Abstract
The antagonism between global energy needs and the obligation to slow global warming is a current challenge. In order to ensure sufficient thermal comfort, the automotive, housing and agricultural building sectors are major energy consumers. Solar control materials and more particularly, selective glazing are part of the solutions proposed to reduce global energy consumption and tackle global warming. In this context, these works are focused on developing new highly ultraviolet (UV) and near-infrared (NIR) absorbent nanocomposite coatings based on K4[{Nb6-xTaxXi12}Xa6]. (X = Cl, Br, 0 ≤ x ≤ 6) transition metal cluster compounds. These compounds contain cluster-based active species that are characterized by their strong absorption of UV and NIR radiations as well as their good transparency in the visible range, which makes them particularly attractive for window applications. Their integration, by solution processes, into a silica-polyethylene glycol or polyvinylpyrrolidone matrices is discussed. Of particular interest is the control and the tuning of their optical properties during the integration and shaping processes. The properties of the solutions and films were investigated by complementary techniques (UV-Vis-NIR spectrometry, ESI-MS, SEM, HRTEM, etc.). Results of these works have led to the development of versatile solar control coatings whose optical properties are competitive with commercialized material.
Collapse
Affiliation(s)
- Clément Lebastard
- Univ Rennes, CNRS, ISCR, UMR6226, F-35000 Rennes, France; (M.W.); (S.C.)
- CNRS-Saint Gobain-NIMS, IRL3629, Laboratory for Innovative Key Materials and Structures (LINK), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Japan; (N.O.); (T.U.)
| | - Maxence Wilmet
- Univ Rennes, CNRS, ISCR, UMR6226, F-35000 Rennes, France; (M.W.); (S.C.)
- CNRS-Saint Gobain-NIMS, IRL3629, Laboratory for Innovative Key Materials and Structures (LINK), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Japan; (N.O.); (T.U.)
- Saint Gobain Research Paris, F-93300 Aubervilliers, France
| | - Stéphane Cordier
- Univ Rennes, CNRS, ISCR, UMR6226, F-35000 Rennes, France; (M.W.); (S.C.)
| | - Clothilde Comby-Zerbino
- Univ Lyon, Univ Claude Bernard Lyon 1, CNRS, Institut Lumière Matière, F-69622 Lyon, France; (C.C.-Z.); (L.M.); (P.D.)
| | - Luke MacAleese
- Univ Lyon, Univ Claude Bernard Lyon 1, CNRS, Institut Lumière Matière, F-69622 Lyon, France; (C.C.-Z.); (L.M.); (P.D.)
| | - Philippe Dugourd
- Univ Lyon, Univ Claude Bernard Lyon 1, CNRS, Institut Lumière Matière, F-69622 Lyon, France; (C.C.-Z.); (L.M.); (P.D.)
| | - Toru Hara
- Research Center for Structural Materials, NIMS, 1-2-1 Sengen, Tsukuba 305-0047, Japan;
| | - Naoki Ohashi
- CNRS-Saint Gobain-NIMS, IRL3629, Laboratory for Innovative Key Materials and Structures (LINK), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Japan; (N.O.); (T.U.)
- Research Center for Functional Materials, NIMS, 1-1 Namiki, Tsukuba 305-0044, Japan
| | - Tetsuo Uchikoshi
- CNRS-Saint Gobain-NIMS, IRL3629, Laboratory for Innovative Key Materials and Structures (LINK), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Japan; (N.O.); (T.U.)
- Research Center for Functional Materials, NIMS, 1-1 Namiki, Tsukuba 305-0044, Japan
| | - Fabien Grasset
- Univ Rennes, CNRS, ISCR, UMR6226, F-35000 Rennes, France; (M.W.); (S.C.)
- CNRS-Saint Gobain-NIMS, IRL3629, Laboratory for Innovative Key Materials and Structures (LINK), National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba 305-0044, Japan; (N.O.); (T.U.)
| |
Collapse
|
39
|
Sharma H, Batra R, Kumar S, Kumar M, Kumar S, Balyan HS, Gupta PK. Identification and characterization of 20S proteasome genes and their relevance to heat/drought tolerance in bread wheat. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
40
|
Linhorst A, Lübke T. The Human Ntn-Hydrolase Superfamily: Structure, Functions and Perspectives. Cells 2022; 11:cells11101592. [PMID: 35626629 PMCID: PMC9140057 DOI: 10.3390/cells11101592] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 01/27/2023] Open
Abstract
N-terminal nucleophile (Ntn)-hydrolases catalyze the cleavage of amide bonds in a variety of macromolecules, including the peptide bond in proteins, the amide bond in N-linked protein glycosylation, and the amide bond linking a fatty acid to sphingosine in complex sphingolipids. Ntn-hydrolases are all sharing two common hallmarks: Firstly, the enzymes are synthesized as inactive precursors that undergo auto-proteolytic self-activation, which, as a consequence, reveals the active site nucleophile at the newly formed N-terminus. Secondly, all Ntn-hydrolases share a structural consistent αββα-fold, notwithstanding the total lack of amino acid sequence homology. In humans, five subclasses of the Ntn-superfamily have been identified so far, comprising relevant members such as the catalytic active subunits of the proteasome or a number of lysosomal hydrolases, which are often associated with lysosomal storage diseases. This review gives an updated overview on the structural, functional, and (patho-)physiological characteristics of human Ntn-hydrolases, in particular.
Collapse
|
41
|
Watanabe A, Yashiroda H, Ishihara S, Lo M, Murata S. The Molecular Mechanisms Governing the Assembly of the Immuno- and Thymoproteasomes in the Presence of Constitutive Proteasomes. Cells 2022; 11:cells11091580. [PMID: 35563886 PMCID: PMC9105311 DOI: 10.3390/cells11091580] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/02/2022] [Accepted: 05/04/2022] [Indexed: 02/06/2023] Open
Abstract
The proteasome is a large protein complex responsible for proteolysis in cells. Though the proteasome is widely conserved in all eukaryotes, vertebrates additionally possess tissue-specific proteasomes, termed immunoproteasomes and thymoproteasomes. These specialized proteasomes diverge from constitutive proteasomes in the makeup of their catalytic 20S core particle (CP), whereby the constitutive β1, β2, and β5 catalytic subunits are replaced by β1i, β2i, and β5i in immunoproteasomes, or β1i, β2i, and β5t in thymoproteasomes. However, as constitutive β1, β2, and β5 are also present in tissues and cells expressing immuno- and thymoproteasomes, the specialized proteasomes must be able to selectively incorporate their specific subunits. Here, we review the mechanisms governing the assembly of constitutive and specialized proteasomes elucidated thus far. Studies have revealed that β1i and β2i are added onto the α-ring of the CP prior to the other β subunits. Furthermore, β5i and β5t can be incorporated independent of β4, whereas constitutive β5 incorporation is dependent on β4. These mechanisms allow the immuno- and thymoproteasomes to integrate tissue-specific β-subunits without contamination from constitutive β1, β2, and β5. We end the review with a brief discussion on the diseases caused by mutations to the immunoproteasome and the proteins involved with its assembly.
Collapse
|
42
|
The YΦ motif defines the structure-activity relationships of human 20S proteasome activators. Nat Commun 2022; 13:1226. [PMID: 35264557 PMCID: PMC8907193 DOI: 10.1038/s41467-022-28864-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 02/11/2022] [Indexed: 11/08/2022] Open
Abstract
The 20S proteasome (20S) facilitates turnover of most eukaryotic proteins. Substrate entry into the 20S first requires opening of gating loops through binding of HbYX motifs that are present at the C-termini of certain proteasome activators (PAs). The HbYX motif has been predominantly characterized in the archaeal 20S, whereas little is known about the sequence preferences of the human 20S (h20S). Here, we synthesize and screen ~120 HbYX-like peptides, revealing unexpected differences from the archaeal system and defining the h20S recognition sequence as the Y-F/Y (YФ) motif. To gain further insight, we create a functional chimera of the optimized sequence, NLSYYT, fused to the model activator, PA26E102A. A cryo-EM structure of PA26E102A-h20S is used to identify key interactions, including non-canonical contacts and gate-opening mechanisms. Finally, we demonstrate that the YФ sequence preferences are tuned by valency, allowing multivalent PAs to sample greater sequence space. These results expand the model for termini-mediated gating and provide a template for the design of h20S activators. The proteasome complexes, composed of 20S core particles and one or two regulatory particles (proteasome activators), degrade most eukaryotic proteins. Here, the authors identify a sequence motif and resolve its interactions mediating the activation of the human 20S proteasome.
Collapse
|
43
|
Schulz L, Sendker FL, Hochberg GKA. Non-adaptive complexity and biochemical function. Curr Opin Struct Biol 2022; 73:102339. [PMID: 35247750 DOI: 10.1016/j.sbi.2022.102339] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/06/2021] [Accepted: 01/24/2022] [Indexed: 11/25/2022]
Abstract
Intricate biochemical structures are usually thought to be useful, because natural selection preserves them from degradation by a constant hail of destructive mutations. Biochemists therefore often deliberately disrupt them to understand how complexity improves protein function or fitness. However, evolutionary theory suggests that even useless complexity that never improved fitness can become completely essential if a simple set of evolutionary conditions is fulfilled. We review evidence that stable protein complexes, protein-chaperone interactions, and complexes consisting of several paralogs all fulfill these conditions. This makes reverse genetics or destructive mutagenesis unsuitable for assigning functions to these kinds of complexity. Instead, we advocate that incorporating evolutionary approaches into biochemistry overcomes this difficulty and allows us to distinguish useless from useful biochemical complexity.
Collapse
Affiliation(s)
- Luca Schulz
- Max Planck Institute for Terrestrial Microbiology, Karl-von-Frisch Straße 10, 35043 Marburg, Germany. https://twitter.com/schulluc
| | - Franziska L Sendker
- Max Planck Institute for Terrestrial Microbiology, Karl-von-Frisch Straße 10, 35043 Marburg, Germany. https://twitter.com/SendkerFL
| | - Georg K A Hochberg
- Max Planck Institute for Terrestrial Microbiology, Karl-von-Frisch Straße 10, 35043 Marburg, Germany; Department of Chemistry, University of Marburg, Hans-Meerwein-Straße 4, 35032 Marburg, Germany; Center for Synthetic Microbiology (SYNMIKRO), Hans-Meerwein-Straße 6, 35032 Marburg, Germany.
| |
Collapse
|
44
|
Functional Differences between Proteasome Subtypes. Cells 2022; 11:cells11030421. [PMID: 35159231 PMCID: PMC8834425 DOI: 10.3390/cells11030421] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/11/2022] [Accepted: 01/14/2022] [Indexed: 12/30/2022] Open
Abstract
Four proteasome subtypes are commonly present in mammalian tissues: standard proteasomes, which contain the standard catalytic subunits β1, β2 and β5; immunoproteasomes containing the immuno-subunits β1i, β2i and β5i; and two intermediate proteasomes, containing a mix of standard and immuno-subunits. Recent studies revealed the expression of two tissue-specific proteasome subtypes in cortical thymic epithelial cells and in testes: thymoproteasomes and spermatoproteasomes. In this review, we describe the mechanisms that enable the ATP- and ubiquitin-dependent as well as the ATP- and ubiquitin-independent degradation of proteins by the proteasome. We focus on understanding the role of the different proteasome subtypes in maintaining protein homeostasis in normal physiological conditions through the ATP- and ubiquitin-dependent degradation of proteins. Additionally, we discuss the role of each proteasome subtype in the ATP- and ubiquitin-independent degradation of disordered proteins. We also discuss the role of the proteasome in the generation of peptides presented by MHC class I molecules and the implication of having different proteasome subtypes for the peptide repertoire presented at the cell surface. Finally, we discuss the role of the immunoproteasome in immune cells and its modulation as a potential therapy for autoimmune diseases.
Collapse
|
45
|
Kisselev AF. Site-Specific Proteasome Inhibitors. Biomolecules 2021; 12:54. [PMID: 35053202 PMCID: PMC8773591 DOI: 10.3390/biom12010054] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [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
|
46
|
Probing allosteric interactions in homo-oligomeric molecular machines using solution NMR spectroscopy. Proc Natl Acad Sci U S A 2021; 118:2116325118. [PMID: 34893543 DOI: 10.1073/pnas.2116325118] [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] [Accepted: 10/27/2021] [Indexed: 11/18/2022] Open
Abstract
Developments in solution NMR spectroscopy have significantly impacted the biological questions that can now be addressed by this methodology. By means of illustration, we present here a perspective focusing on studies of a number of molecular machines that are critical for cellular homeostasis. The role of NMR in elucidating the structural dynamics of these important molecules is emphasized, focusing specifically on intersubunit allosteric communication in homo-oligomers. In many biophysical studies of oligomers, allostery is inferred by showing that models specifically including intersubunit communication best fit the data of interest. Ideally, however, experimental studies focusing on one subunit of a multisubunit system would be performed as an important complement to the more traditional bulk measurements in which signals from all components are measured simultaneously. Using an approach whereby asymmetric molecules are prepared in concert with NMR experiments focusing on the structural dynamics of individual protomers, we present examples of how intersubunit allostery can be directly observed in high-molecular-weight protein systems. These examples highlight some of the unique roles of solution NMR spectroscopy in studies of complex biomolecules and emphasize the important synergy between NMR and other atomic resolution biophysical methods.
Collapse
|
47
|
Zhang S, Huang G, Versloot R, Bruininks BMH, Telles de Souza PC, Marrink SJ, Maglia G. Bottom-up fabrication of a proteasome-nanopore that unravels and processes single proteins. Nat Chem 2021; 13:1192-1199. [PMID: 34795436 PMCID: PMC7612055 DOI: 10.1038/s41557-021-00824-w] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 09/22/2021] [Indexed: 02/08/2023]
Abstract
The precise assembly and engineering of molecular machines capable of handling biomolecules play crucial roles in most single-molecule methods. In this work we use components from all three domains of life to fabricate an integrated multiprotein complex that controls the unfolding and threading of individual proteins across a nanopore. This 900 kDa multicomponent device was made in two steps. First, we designed a stable and low-noise β-barrel nanopore sensor by linking the transmembrane region of bacterial protective antigen to a mammalian proteasome activator. An archaeal 20S proteasome was then built into the artificial nanopore to control the unfolding and linearized transport of proteins across the nanopore. This multicomponent molecular machine opens the door to two approaches in single-molecule protein analysis, in which selected substrate proteins are unfolded, fed to into the proteasomal chamber and then addressed either as fragmented peptides or intact polypeptides.
Collapse
|
48
|
PDB-wide identification of physiological hetero-oligomeric assemblies based on conserved quaternary structure geometry. Structure 2021; 29:1303-1311.e3. [PMID: 34520740 PMCID: PMC8575123 DOI: 10.1016/j.str.2021.07.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 03/22/2021] [Accepted: 07/23/2021] [Indexed: 11/21/2022]
Abstract
An accurate understanding of biomolecular mechanisms and diseases requires information on protein quaternary structure (QS). A critical challenge in inferring QS information from crystallography data is distinguishing biological interfaces from fortuitous crystal-packing contacts. Here, we employ QS conservation across homologs to infer the biological relevance of hetero-oligomers. We compare the structures and compositions of hetero-oligomers, which allow us to annotate 7,810 complexes as physiologically relevant, 1,060 as likely errors, and 1,432 with comparative information on subunit stoichiometry and composition. Excluding immunoglobulins, these annotations encompass over 51% of hetero-oligomers in the PDB. We curate a dataset of 577 hetero-oligomeric complexes to benchmark these annotations, which reveals an accuracy >94%. When homology information is not available, we compare QS across repositories (PDB, PISA, and EPPIC) to derive confidence estimates. This work provides high-quality annotations along with a large benchmark dataset of hetero-assemblies.
Collapse
|
49
|
Bonea D, Noureddine J, Gazzarrini S, Zhao R. Oxidative and salt stresses alter the 26S proteasome holoenzyme and associated protein profiles in Arabidopsis thaliana. BMC PLANT BIOLOGY 2021; 21:486. [PMID: 34696730 PMCID: PMC8543921 DOI: 10.1186/s12870-021-03234-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/29/2021] [Indexed: 05/13/2023]
Abstract
BACKGROUND The 26S proteasome, canonically composed of multi-subunit 19S regulatory (RP) and 20S core (CP) particles, is crucial for cellular proteostasis. Proteasomes are re-modeled, activated, or re-localized and this regulation is critical for plants in response to environmental stresses. The proteasome holoenzyme assembly and dissociation are therefore highly dynamic in vivo. However, the stoichiometric changes of the plant proteasomes and how proteasome associated chaperones vary under common abiotic stresses have not been systematically studied. RESULTS Here, we studied the impact of abiotic stresses on proteasome structure, activity, and interacting partners in Arabidopsis thaliana. We analyzed available RNA expression data and observed that expressions of proteasome coding genes varied substantially under stresses; however, the protein levels of a few key subunits did not change significantly within 24 h. Instead, a switch in the predominant proteasome complex, from 26S to 20S, occurs under oxidative or salt stress. Oxidative stress also reduced the cellular ATP content and the association of HSP70-family proteins to the 20S proteasome, but enhanced the activity of cellular free form CP. Salt stress, on the other hand, did not affect cellular ATP level, but caused subtle changes in proteasome subunit composition and impacted bindings of assembly chaperones. Analyses of an array of T-DNA insertional mutant lines highlighted important roles for several putative assembly chaperones in seedling establishment and stress sensitivity. We also observed that knockout of PBAC1, one of the α-ring assembly chaperones, resulted in reduced germination and tearing of the seed coat following sterilization. CONCLUSIONS Our study revealed an evolutionarily conserved mechanism of proteasome regulation during oxidative stress, involving dynamic regulation of the holoenzyme formation and associated regulatory proteins, and we also identified a novel role of the PBAC1 proteasome assembly chaperone in seed coat development.
Collapse
Affiliation(s)
- Diana Bonea
- Department of Biological Sciences, University of Toronto, 1265 Military Trail, Toronto, ON M1C 1A4 Canada
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5 Canada
| | - Jenan Noureddine
- Department of Biological Sciences, University of Toronto, 1265 Military Trail, Toronto, ON M1C 1A4 Canada
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5 Canada
| | - Sonia Gazzarrini
- Department of Biological Sciences, University of Toronto, 1265 Military Trail, Toronto, ON M1C 1A4 Canada
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5 Canada
| | - Rongmin Zhao
- Department of Biological Sciences, University of Toronto, 1265 Military Trail, Toronto, ON M1C 1A4 Canada
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5 Canada
| |
Collapse
|
50
|
Dong S, Chen H, Zhou Q, Liao N. Protein degradation control and regulation of bacterial survival and pathogenicity: the role of protein degradation systems in bacteria. Mol Biol Rep 2021; 48:7575-7585. [PMID: 34655017 DOI: 10.1007/s11033-021-06744-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/01/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Protein degradation systems play crucial roles in all the kingdoms of life. Their natural function is to eliminate proteins that are improperly synthesized, damaged, aggregated, or short-lived, ensuring the timely and accurate regulation of the response to abrupt environmental changes. Thus, proteolysis plays an important role in protein homeostasis, quality control, and the control of regulatory processes, such as adaptation and cell development. Except for the lysosome, ATPases Associated with various cellular Activities (AAA+) ATPase-protease complex is another major protein degradation system in the cell. METHODS AND RESULTS The AAA+ ATPase-protease complex is a giant energy-dependent protease complex found in almost all kinds of cells, including bacteria, archaea and eukarya. Based on sequence analysis of ClpQ (HslV) and 20S proteasome beta subunits, it was found that bacterial ClpQ possess multiple same highly conserved motifs with 20S proteasome beta subunits of archaea and eukaryote. In this review, we also discussed the structure and functional mechanism, protein degradation signals and pathogenic role of proteasome / Clp protease complex in prokaryotes. CONCLUSION Bacterial protein degradation systems play important roles in stress tolerance, protein quality control, DNA protection, transcription and pathogenicity of bacteria. But our current knowledge of the bacterial protease system is incomplete, and further research into the Clp protease complex and associated protein degradation signals will extend our understanding of the metabolism, physiology, reproduction, and pathogenicity of bacteria.
Collapse
Affiliation(s)
- Shilei Dong
- Department of Clinical Laboratory, Zhejiang Hospital, Hangzhou, 310013, China
| | - Honghu Chen
- Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, 310051, China
| | - Qingxue Zhou
- Department of Clinical Laboratory, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, 310008, China
| | - Ningbo Liao
- College of Food Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China.
| |
Collapse
|