1
|
Santoro AM, Persico M, D’Urso A, Cunsolo A, Tkachuk O, Milardi D, Purrello R, Tundo GR, Sbardella D, Osmulski PA, Gaczynska M, Coletta M, Fattorusso C. Tetra-anionic porphyrin mimics protein-protein interactions between regulatory particles and the catalytic core, allosterically activating human 20S proteasome. J Enzyme Inhib Med Chem 2025; 40:2482892. [PMID: 40192126 PMCID: PMC11980194 DOI: 10.1080/14756366.2025.2482892] [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: 10/09/2024] [Revised: 03/03/2025] [Accepted: 03/11/2025] [Indexed: 04/11/2025] Open
Abstract
Decreased proteasome activity is a hallmark of brain and retinal neurodegenerative diseases (Alzheimer's, Parkinson's diseases, glaucoma) boosting the search for molecules acting as proteasome activators. Based on the hypothesis of an electrostatic key code driving catalytic core particle (20S) activation by regulatory particles (RPs), we identified the tetra-anionic meso-Tetrakis(4-sulphonatophenyl)-porphyrin (H2TPPS) as a new activator of human proteasome. By means of an integrated approach, including bioinformatics, enzymatic kinetic analysis, atomic force microscopy, and dynamic docking simulations, we show how binding of H2TPPS affects the closed/open conformational equilibrium of human 20S to ultimately promote substrate gate opening and proteolytic activity. These outcomes support our hypothesis and pave the way to the rational discovery of new proteasome allosteric modulators able to reproduce the key structural elements of regulatory particles responsible for catalytic activation.
Collapse
Affiliation(s)
- A. M. Santoro
- National Research Council, Institute of Crystallography, Sede Secondaria di Catania, Catania, Italy
| | - M. Persico
- Department of Pharmacy, University of Naples “Federico II”, Napoli, Italy
| | - A. D’Urso
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - A. Cunsolo
- Department of Chemical Sciences, University of Catania, Catania, Italy
- Epic Sciences, San Diego, California, USA
| | - O. Tkachuk
- Department of Pharmacy, University of Naples “Federico II”, Napoli, Italy
| | - D. Milardi
- National Research Council, Institute of Crystallography, Sede Secondaria di Catania, Catania, Italy
| | - R. Purrello
- Department of Chemical Sciences, University of Catania, Catania, Italy
| | - G. R. Tundo
- Department of Clinical Sciences and Translational Medicine, University of Roma Tor Vergata, Roma, Italy
| | | | - P. A. Osmulski
- Department of Molecular Medicine, University of Texas Health at San Antonio, San Antonio, Texas, USA
| | - M. Gaczynska
- Department of Molecular Medicine, University of Texas Health at San Antonio, San Antonio, Texas, USA
| | | | - C. Fattorusso
- Department of Pharmacy, University of Naples “Federico II”, Napoli, Italy
| |
Collapse
|
2
|
Theocharaki K, Anastasiadi AT, Delicou S, Tzounakas VL, Barla I, Rouvela S, Kazolia E, Tzafa G, Mpekoulis G, Gousdovas T, Pavlou E, Kostopoulos IV, Velentzas AD, Simantiris N, Xydaki A, Vassilaki N, Voskaridou E, Aggeli IK, Nomikou E, Tsitsilonis O, Papageorgiou E, Thomaidis N, Gikas E, Politou M, Komninaka V, Antonelou MH. Cellular and biochemical heterogeneity contributes to the phenotypic diversity of transfusion-dependent β-thalassemia. Blood Adv 2025; 9:2091-2107. [PMID: 39928952 DOI: 10.1182/bloodadvances.2024015232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/13/2025] [Accepted: 01/29/2025] [Indexed: 02/12/2025] Open
Abstract
ABSTRACT Transfusion-dependent thalassemia (TDT) is a type of protein aggregation disease. Its clinical heterogeneity imposes challenges in effective management. Red blood cell (RBC) variables may be clinically relevant as mechanistic parts or tellers of TDT pathophysiology. This is a cross-sectional study of RBC and plasma physiology in adult patients with TDT vs healthy control. TDT plasma was characterized by increased protein carbonylation, antioxidants, and larger than normal extracellular vesicles. RBCs were osmotically resistant but prone to oxidative hemolysis. They overexposed phosphatidylserine and exhibited pathologically low proteasome proteolytic activity (PPA), which correlated with metabolic markers of the disease. RBC ultrastructure was distorted, with splenectomy-related membrane pits of 300 to 800 nm. Plasma metabolomics revealed differences in heme metabolism, redox potential, short-chain fatty acids, and nitric oxide bioavailability, but also in catecholamine pathways. According to coefficient of variation assessment, hemolysis, iron homeostasis, PPA, and phosphatidylserine exposure were highly variable among patients, as opposed to RBC fragility and plasma antioxidants, amino acids, and catecholamines. Sex-based differences were detected in hemolysis, redox, and energy variables, whereas splenectomy-related differences referred to thrombotic risk, RBC morphology, and plasma metabolites with neuroendocrine activity. Hepcidin varied according to oxidative hemolysis and metabolic markers of bacterial activity. Patients with higher pretransfusion hemoglobin levels (>10 g/dL) presented mildly distorted profiles and lower membrane-associated PPA, whereas classification by severity of mutations revealed different levels of hemostasis, inflammation, plasma epinephrine, hexosamines, and methyltransferase activity markers. The currently reported heterogeneity of cellular and biochemical features probably contributes to the wide phenotypic diversity of TDT at clinical level.
Collapse
Affiliation(s)
- Konstantina Theocharaki
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Alkmini T Anastasiadi
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Sophia Delicou
- Thalassemia and Sickle Cell Unit, Expertise Center of Hemoglobinopathies and their Complications, Hippokration General Hospital of Athens, Athens, Greece
| | - Vassilis L Tzounakas
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Barla
- Department of Chemistry, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Stella Rouvela
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Evgenia Kazolia
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgia Tzafa
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - George Mpekoulis
- Department of Microbiology, Laboratory of Molecular Virology, Hellenic Pasteur Institute, Athens, Greece
| | - Theodore Gousdovas
- Hematology Laboratory-Blood Bank, Aretaieio Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Efthymia Pavlou
- Blood Bank and Haemophilia Unit, Hippokration General Hospital of Athens, Athens, Greece
| | - Ioannis V Kostopoulos
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanassios D Velentzas
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Aikaterini Xydaki
- Thalassemia and Sickle Cell Unit, Expertise Center of Hemoglobinopathies and their Complications, Hippokration General Hospital of Athens, Athens, Greece
| | - Niki Vassilaki
- Department of Microbiology, Laboratory of Molecular Virology, Hellenic Pasteur Institute, Athens, Greece
| | - Ersi Voskaridou
- Centre of Excellence in Rare Haematological (Haemoglobinopathies) & Rare Metabolic (Gaucher Disease) Diseases, Laiko General Hospital, Athens, Greece
| | - Ioanna-Katerina Aggeli
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Efrosyni Nomikou
- Blood Bank and Haemophilia Unit, Hippokration General Hospital of Athens, Athens, Greece
| | - Ourania Tsitsilonis
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathia Papageorgiou
- Department of Biomedical Sciences, School of Health & Caring Sciences, University of West Attica, Egaleo, Greece
| | - Nikolaos Thomaidis
- Department of Chemistry, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Gikas
- Department of Chemistry, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| | - Marianna Politou
- Hematology Laboratory-Blood Bank, Aretaieio Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Veroniki Komninaka
- Centre of Excellence in Rare Haematological (Haemoglobinopathies) & Rare Metabolic (Gaucher Disease) Diseases, Laiko General Hospital, Athens, Greece
| | - Marianna H Antonelou
- Department of Biology, School of Science, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
3
|
Sowik D, Giżyńska M, Trepczyk K, Witkowska J, Jankowska E. Activation of the 20S proteasome as a possible strategy to counteract amylin oligomerization in type 2 diabetes. Bioorg Chem 2025; 155:108157. [PMID: 39809118 DOI: 10.1016/j.bioorg.2025.108157] [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: 10/17/2024] [Revised: 12/19/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Human amylin, called also islet amyloid polypeptide (hIAPP), is the principal constituent of amyloid deposits in the pancreatic islets. Together with hyperglycemia, hIAPP-derived oligomers and aggregates are important culprits in type 2 diabetes mellitus (T2DM). Preventing aggregation, and in particular inhibiting the formation and/or stimulating degradation of toxic amylin oligomers formed early in the process, may reduce the negative effects of T2DM. Such therapeutic intervention may be enabled by activation of the 20S proteasome, a proteolytic system responsible for digesting proteins that are damaged or natively exhibit aggregation tendencies. In this work, we showed that in the lag phase of the aggregation process, soluble oligomers of small size (dimer to heptamer) were present alongside the amylin monomer. These oligomers inhibited the activity of the human 20S proteasome (h20S). To counteract this inhibition, we designed two activators that proved to be effective in restoring the peptidase efficiency of h20S to basal levels and even stimulating the enzyme to degrade the fluorogenic substrate more efficiently. They showed this effect both against isolated h20S and in cell lysate.
Collapse
Affiliation(s)
- Daria Sowik
- Department of Biomedical Chemistry Faculty of Chemistry University of Gdansk Wita Stwosza 63 Gdańsk Poland
| | - Małgorzata Giżyńska
- Department of Biomedical Chemistry Faculty of Chemistry University of Gdansk Wita Stwosza 63 Gdańsk Poland
| | - Karolina Trepczyk
- Department of Biomedical Chemistry Faculty of Chemistry University of Gdansk Wita Stwosza 63 Gdańsk Poland
| | - Julia Witkowska
- Department of Biomedical Chemistry Faculty of Chemistry University of Gdansk Wita Stwosza 63 Gdańsk Poland
| | - Elżbieta Jankowska
- Department of Biomedical Chemistry Faculty of Chemistry University of Gdansk Wita Stwosza 63 Gdańsk Poland.
| |
Collapse
|
4
|
Arkinson C, Dong KC, Gee CL, Martin A. Mechanisms and regulation of substrate degradation by the 26S proteasome. Nat Rev Mol Cell Biol 2025; 26:104-122. [PMID: 39362999 PMCID: PMC11772106 DOI: 10.1038/s41580-024-00778-0] [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] [Accepted: 08/23/2024] [Indexed: 10/05/2024]
Abstract
The 26S proteasome is involved in degrading and regulating the majority of proteins in eukaryotic cells, which requires a sophisticated balance of specificity and promiscuity. In this Review, we discuss the principles that underly substrate recognition and ATP-dependent degradation by the proteasome. We focus on recent insights into the mechanisms of conventional ubiquitin-dependent and ubiquitin-independent protein turnover, and discuss the plethora of modulators for proteasome function, including substrate-delivering cofactors, ubiquitin ligases and deubiquitinases that enable the targeting of a highly diverse substrate pool. Furthermore, we summarize recent progress in our understanding of substrate processing upstream of the 26S proteasome by the p97 protein unfoldase. The advances in our knowledge of proteasome structure, function and regulation also inform new strategies for specific inhibition or harnessing the degradation capabilities of the proteasome for the treatment of human diseases, for instance, by using proteolysis targeting chimera molecules or molecular glues.
Collapse
Affiliation(s)
- Connor Arkinson
- California Institute for Quantitative Biosciences, University of California at Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA, USA
| | - Ken C Dong
- Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA, USA
| | - Christine L Gee
- California Institute for Quantitative Biosciences, University of California at Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA, USA
| | - Andreas Martin
- California Institute for Quantitative Biosciences, University of California at Berkeley, Berkeley, CA, USA.
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA.
- Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA, USA.
| |
Collapse
|
5
|
Sahin F, Gunel A, Atasoy BT, Guler U, Salih B, Kuzu I, Taspinar M, Cinar O, Kahveci S. Enhancing proteasome activity by NMDAR antagonists explains their therapeutic effect in neurodegenerative and mental diseases. Sci Rep 2025; 15:1165. [PMID: 39805913 PMCID: PMC11729902 DOI: 10.1038/s41598-024-84479-w] [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: 08/25/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025] Open
Abstract
NMDAR antagonists, such as memantine and ketamine, have shown efficacy in treating neurodegenerative diseases and major depression. The mechanism by which these drugs correct the aforementioned diseases is still unknown. Our study reveals that these antagonists significantly enhance 20S proteasome activity, crucial for degrading intrinsically disordered, oxidatively damaged, or misfolded proteins, factors pivotal in neurodegenerative diseases like Alzheimer's and Parkinson's. In our mouse model experiment, ketamine administration notably altered brain synaptic protein profiles within two hours, significantly downregulating proteins strongly associated with Alzheimer's and Parkinson's diseases. Furthermore, the altered proteins exhibited enrichment in terms related to plasticity and potentiation, including retrograde endocannabinoid signaling-a pivotal pathway in both short- and long-term plasticity that may elucidate the long-lasting effects of ketamine in major depression. Via the ubiquitin-independent 20S proteasome pathway (UIPS), these drugs maintain cellular protein homeostasis, which is crucial as proteasome activity declines with age, leading to protein aggregation and disease symptoms. Therefore, these findings hold promise for new treatment options not only for brain diseases but also for other systemic conditions associated with unfolded or misfolded proteins.
Collapse
Affiliation(s)
- Fikret Sahin
- Department of Medical Microbiology, Ankara University School of Medicine, Ankara, Turkey.
| | - Aslihan Gunel
- Faculty of Arts and Science Department of Chemistry-Biochemistry, Kırşehir Ahi Evran University, Kırşehir, Turkey
| | - Buse Turegun Atasoy
- Department of Medical Microbiology, Ankara University School of Medicine, Ankara, Turkey
| | - Ulku Guler
- Department of Chemistry, Hacettepe University, Ankara, Turkey
| | - Bekir Salih
- Department of Chemistry, Hacettepe University, Ankara, Turkey
| | - Isinsu Kuzu
- Department of Medical Pathology, Ankara University School of Medicine, Ankara, Turkey
| | - Mehmet Taspinar
- Department of Medical Biology, Aksaray University School of Medicine, Aksaray, Turkey
| | - Ozgur Cinar
- Department of Histology and Embryology, Ankara University School of Medicine, Ankara, Turkey
| | - Selda Kahveci
- Department of Histology and Embryology, Ankara University School of Medicine, Ankara, Turkey
- Department of Histology and Embryology, Yozgat Bozok University, Yozgat, Turkey
| |
Collapse
|
6
|
Kim H, Kim H, Lee SH, Kwon JH, Byun S, Yoo JY, Park SY, Yoon HG. Deubiquitinase inhibitor bAP-15 suppresses renal epithelial to mesenchymal transition via inhibition of p300 stability. Biochem Biophys Res Commun 2024; 741:151095. [PMID: 39622158 DOI: 10.1016/j.bbrc.2024.151095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/11/2024]
Abstract
Renal fibrosis is an irreversible disease that is common in patients with chronic kidney disease. Elevated levels of the histone acetyltransferase p300 have been reported in various fibrotic diseases, including renal fibrosis, suggesting that p300 may be a promising therapeutic target. To investigate the specific deubiquitinase (DUB) involved in the regulation of p300 protein stability in renal epithelial cells, we tested 13 DUB inhibitors using a kidney tubular epithelial cell line. We found that the p300-specific DUB inhibitor, bAP-15 reduces p300 protein stability by targeting ubiquitin-specific protease 14 (USP14) and ubiquitin C-terminal hydrolase L5 (UCHL5). The mRNA levels of USP14 and UCHL5 were increased in patients with chronic kidney disease, and increased protein levels of USP14 and UCHL5 during fibrosis progression were validated using a mouse renal fibrosis model. Both USP14 and UCHL5 interacted with p300 in kidney tubular epithelial cells, with increased binding affinity in response to TGF-β. Moreover, bAP-15-induced p300 degradation inhibited epithelial-to-mesenchymal transition and reduced the expression of pro-fibrotic target genes. Our findings demonstrate an anti-fibrotic effect of bAP-15 through the regulation of p300 stability and suggest that bAP-15 may be a potential therapeutic agent for renal fibrosis.
Collapse
Affiliation(s)
- Hyunsik Kim
- Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Hyunseung Kim
- Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Sun-Ho Lee
- Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Jae-Hwan Kwon
- Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Seunghee Byun
- Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Jung-Yoon Yoo
- Department of Biomedical Laboratory Science, Yonsei University MIRAE Campus, Wonju, 26493, South Korea
| | - Soo-Yeon Park
- Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| | - Ho-Geun Yoon
- Department of Biochemistry and Molecular Biology, Severance Medical Research Institute, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
7
|
Desouky MA, Michel HE, Elsherbiny DA, George MY. Recent pharmacological insights on abating toxic protein species burden in neurological disorders: Emphasis on 26S proteasome activation. Life Sci 2024; 359:123206. [PMID: 39489397 DOI: 10.1016/j.lfs.2024.123206] [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: 06/08/2024] [Revised: 07/30/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Protein homeostasis (proteostasis) refers to the plethora of mechanisms that safeguard the proper folding of the newly synthesized proteins. It entails various intricately regulated cues that demolish the toxic protein species to prevent their aggregation. The ubiquitin-proteasome system (UPS) is recognized as a salient protein degradation system, with a substantial role in maintaining proteostasis. However, under certain circumstances the protein degradation capacity of the UPS is overwhelmed, leading to the accumulation of misfolded proteins. Several neurodegenerative disorders, such as Alzheimer's disease, Parkinson's disease, Huntington disease, and amyotrophic lateral sclerosis are characterized with the presence of protein aggregates and proteinopathy. Accordingly, enhancing the 26S proteasome degradation activity might delineate a pioneering approach in targeting various proteotoxic disorders. Regrettably, the exact molecular approaches that enhance the proteasomal activity are still not fully understood. Therefore, this review aimed to underscore several signaling cascades that might restore the degradation capacity of this molecular machine. In this review, we discuss the different molecular components of the UPS and how 26S proteasomes are deleteriously affected in many neurodegenerative diseases. Moreover, we summarize different signaling pathways that can be utilized to renovate the 26S proteasome functional capacity, alongside currently known druggable targets in this circuit and various classes of proteasome activators.
Collapse
Affiliation(s)
- Mahmoud A Desouky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Haidy E Michel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Doaa A Elsherbiny
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, 11566 Cairo, Egypt.
| |
Collapse
|
8
|
Liu J, Zhou J, You C, Xia H, Gao Y, Liu Y, Gong X. Research progress in the mechanism of acupuncture regulating microglia in the treatment of Alzheimer's disease. Front Neurosci 2024; 18:1435082. [PMID: 39145293 PMCID: PMC11321967 DOI: 10.3389/fnins.2024.1435082] [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: 05/19/2024] [Accepted: 07/22/2024] [Indexed: 08/16/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease in the central nervous system, characterized by memory and cognitive dysfunction. Acupuncture is an effective means to alleviate the symptoms of AD. Recent studies have shown that microglia play an important role in the occurrence and development of AD. Acupuncture can regulate the activity of microglia, inhibit neuroinflammation, regulate phagocytosis, and clear Aβ Pathological products such as plaque can protect nerve cells and improve cognitive function in AD patients. This article summarizes the relationship between microglia and AD, as well as the research progress in the mechanism of acupuncture regulating microglia in the treatment of AD. The mechanism of acupuncture regulating microglia in the treatment of AD is mainly reviewed from two aspects: inhibiting neuroinflammatory activity and regulating phagocytic function.
Collapse
Affiliation(s)
- Jia Liu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Dalian Medical University College of Integrated Traditional Chinese and Western Medicine, Dalian, China
| | - Jiaqi Zhou
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Chong You
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Dalian Medical University College of Integrated Traditional Chinese and Western Medicine, Dalian, China
| | - Haonan Xia
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Dalian Medical University College of Integrated Traditional Chinese and Western Medicine, Dalian, China
| | - Yuling Gao
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yong Liu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaoyang Gong
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
9
|
McDade E, Liu H, Bui Q, Hassenstab J, Gordon B, Benzinger T, Shen Y, Timsina J, Wang L, Sung YJ, Karch C, Renton A, Daniels A, Morris J, Xiong C, Ibanez L, Perrin R, Llibre-Guerra JJ, Day G, Supnet-Bell C, Xu X, Berman S, Chhatwal J, Ikeuchi T, Kasuga K, Niimi Y, Huey E, Schofield P, Brooks W, Ryan N, Jucker M, Laske C, Levin J, Vöglein J, Roh JH, Lopera F, Bateman R, Cruchaga C. Ubiquitin-Proteasome System in the Different Stages of Dominantly Inherited Alzheimer's Disease. RESEARCH SQUARE 2024:rs.3.rs-4202125. [PMID: 39108475 PMCID: PMC11302696 DOI: 10.21203/rs.3.rs-4202125/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
This study explored the role of the ubiquitin-proteasome system (UPS) in dominantly inherited Alzheimer's disease (DIAD) by examining changes in cerebrospinal fluid (CSF) levels of UPS proteins along with disease progression, AD imaging biomarkers (PiB PET, tau PET), neurodegeneration imaging measures (MRI, FDG PET), and Clinical Dementia Rating® (CDR®). Using the SOMAscan assay, we detected subtle increases in specific ubiquitin enzymes associated with proteostasis in mutation carriers (MCs) up to two decades before the estimated symptom onset. This was followed by more pronounced elevations of UPS-activating enzymes, including E2 and E3 proteins, and ubiquitin-related modifiers. Our findings also demonstrated consistent correlations between UPS proteins and CSF biomarkers such as Aβ42/40 ratio, total tau, various phosphorylated tau species to total tau ratios (ptau181/T181, ptauT205/T205, ptauS202/S202, ptauT217/T217), and MTBR-tau243, alongside Neurofilament light chain (NfL) and the CDR®. Notably, a positive association was observed with imaging markers (PiB PET, tau PET) and a negative correlation with markers of neurodegeneration (FDG PET, MRI), highlighting a significant link between UPS dysregulation and neurodegenerative processes. The correlations suggest that the increase in multiple UPS proteins with rising tau levels and tau-tangle associated markers, indicating a potential role for the UPS in relation to misfolded tau/neurofibrillary tangles (NFTs) and symptom onset. These findings indicate that elevated CSF UPS proteins in DIAD MCs could serve as early indicators of disease progression and suggest a link between UPS dysregulation and amyloid plaque, tau tangles formation, implicating the UPS as a potential therapeutic target in AD pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Alan Renton
- Nash Family Department of Neuroscience and Ronald Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA: Departments of Neurology and Genetics and Ge
| | | | | | | | | | | | | | | | | | | | | | - Jasmeer Chhatwal
- Massachusetts General Hospital, Brigham and Women's Hospital, Harvard Medical School
| | | | - Kensaku Kasuga
- Department of Molecular Genetics, Brain Research Institute, Niigata University
| | | | | | | | | | | | | | | | | | | | | | | | - Randall Bateman
- Department of Neurology, Washington University School of Medicine
| | | |
Collapse
|
10
|
Guo X, Mutch M, Torres AY, Nano M, Rauth N, Harwood J, McDonald D, Chen Z, Montell C, Dai W, Montell DJ. The Zn 2+ transporter ZIP7 enhances endoplasmic-reticulum-associated protein degradation and prevents neurodegeneration in Drosophila. Dev Cell 2024; 59:1655-1667.e6. [PMID: 38670102 PMCID: PMC11233247 DOI: 10.1016/j.devcel.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 12/15/2023] [Accepted: 04/03/2024] [Indexed: 04/28/2024]
Abstract
Proteotoxic stress drives numerous degenerative diseases. Cells initially adapt to misfolded proteins by activating the unfolded protein response (UPR), including endoplasmic-reticulum-associated protein degradation (ERAD). However, persistent stress triggers apoptosis. Enhancing ERAD is a promising therapeutic approach for protein misfolding diseases. The ER-localized Zn2+ transporter ZIP7 is conserved from plants to humans and required for intestinal self-renewal, Notch signaling, cell motility, and survival. However, a unifying mechanism underlying these diverse phenotypes was unknown. In studying Drosophila border cell migration, we discovered that ZIP7-mediated Zn2+ transport enhances the obligatory deubiquitination of proteins by the Rpn11 Zn2+ metalloproteinase in the proteasome lid. In human cells, ZIP7 and Zn2+ are limiting for deubiquitination. In a Drosophila model of neurodegeneration caused by misfolded rhodopsin (Rh1), ZIP7 overexpression degrades misfolded Rh1 and rescues photoreceptor viability and fly vision. Thus, ZIP7-mediated Zn2+ transport is a previously unknown, rate-limiting step for ERAD in vivo with therapeutic potential in protein misfolding diseases.
Collapse
Affiliation(s)
- Xiaoran Guo
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110, USA
| | - Morgan Mutch
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110, USA
| | - Alba Yurani Torres
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110, USA
| | - Maddalena Nano
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110, USA
| | - Nishi Rauth
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110, USA
| | - Jacob Harwood
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110, USA
| | - Drew McDonald
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110, USA
| | - Zijing Chen
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110, USA
| | - Craig Montell
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110, USA
| | - Wei Dai
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110, USA; Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China.
| | - Denise J Montell
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110, USA.
| |
Collapse
|
11
|
Bühringer H, Doroudgar S, Wang X, Frey N, Rangrez AY. Editorial: Proteostasis in cardiac health and disease. Front Mol Biosci 2024; 11:1433721. [PMID: 38882607 PMCID: PMC11176937 DOI: 10.3389/fmolb.2024.1433721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 06/18/2024] Open
Affiliation(s)
- Hannah Bühringer
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg/Mannheim, Germany
| | - Shirin Doroudgar
- Department of Internal Medicine and the Translational Cardiovascular Research Center, University of Arizona College of Medicine–Phoenix, Phoenix, AZ, United States
| | - Xuejun Wang
- Division of Basic Biomedical Sciences, University of South Dakota Sanford School of Medicine, Vermillion, SD, United States
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg/Mannheim, Germany
| | - Ashraf Yusuf Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Heidelberg/Mannheim, Germany
| |
Collapse
|
12
|
Yu Q, Wang Z, Tu Y, Cao Y, Zhu H, Shao J, Zhuang R, Zhou Y, Zhang J. Proteasome activation: A novel strategy for targeting undruggable intrinsically disordered proteins. Bioorg Chem 2024; 145:107217. [PMID: 38368657 DOI: 10.1016/j.bioorg.2024.107217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/23/2024] [Accepted: 02/14/2024] [Indexed: 02/20/2024]
Abstract
Intrinsically disordered proteins (IDPs) are characterized by their inability to adopt well-defined tertiary structures under physiological conditions. Nonetheless, they often play pivotal roles in the progression of various diseases, including cancer, neurodegenerative disorders, and cardiovascular ailments. Owing to their inherent dynamism, conventional drug design approaches based on structural considerations encounter substantial challenges when applied to IDPs. Consequently, the pursuit of therapeutic interventions directed towards IDPs presents a complex endeavor. While there are indeed existing methodologies for targeting IDPs, they are encumbered by noteworthy constrains. Hence, there exists an imminent imperative to investigate more efficacious and universally applicable strategies for modulating IDPs. Here, we present an overview of the latest advancements in the research pertaining to IDPs, along with the indirect regulation approach involving the modulation of IDP degradation through proteasome. By comprehending these advancements in research, novel insights can be generated to facilitate the development of new drugs targeted at addressing the accumulation of IDPs in diverse pathological conditions.
Collapse
Affiliation(s)
- Qian Yu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang Province, China; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China
| | - Zheng Wang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang Province, China; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China
| | - Yutong Tu
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yu Cao
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, Zhejiang Province, China
| | - Huajian Zhu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang Province, China; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China
| | - Jiaan Shao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang Province, China; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China
| | - Rangxiao Zhuang
- Department of Pharmaceutical Preparation, Hangzhou Xixi Hospital, Hangzhou, 310023, Zhejiang Province, China.
| | - Yubo Zhou
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Jiankang Zhang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang Province, China; College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang Province, China.
| |
Collapse
|
13
|
Amrein Almira A, Chen MW, El Demerdash N, Javdan C, Park D, Lee JK, Martin LJ. Proteasome localization and activity in pig brain and in vivo small molecule screening for activators. Front Cell Neurosci 2024; 18:1353542. [PMID: 38469354 PMCID: PMC10925635 DOI: 10.3389/fncel.2024.1353542] [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: 12/10/2023] [Accepted: 02/06/2024] [Indexed: 03/13/2024] Open
Abstract
Introduction Loss of proteasome function, proteinopathy, and proteotoxicity may cause neurodegeneration across the human lifespan in several forms of brain injury and disease. Drugs that activate brain proteasomes in vivo could thus have a broad therapeutic impact in neurology. Methods Using pigs, a clinically relevant large animal with a functionally compartmental gyrencephalic cerebral cortex, we evaluated the localization and biochemical activity of brain proteasomes and tested the ability of small molecules to activate brain proteasomes. Results By Western blotting, proteasome protein subunit PSMB5 and PSMA3 levels were similar in different pig brain regions. Immunohistochemistry for PSMB5 showed localization in the cytoplasm (diffuse and particulate) and nucleus (cytoplasm < nucleus). Some PSMB5 immunoreactivity was colocalized with mitochondrial (voltage-gated anion channel and cyclophilin D) and cell death (Aven) proteins in the neuronal soma and neuropil in the neocortex of pig and human brains. In the nucleus, PSMB5 immunoreactivity was diffuse, particulate, and clustered, including perinucleolar decorations. By fluorogenic assay, proteasome chymotrypsin-like activities (CTL) in crude tissue soluble fractions were generally similar within eight different pig brain regions. Proteasome CTL activity in the hippocampus was correlated with activity in nasal mucosa biopsies. In pilot analyses of subcellular fractions of pig cerebral cortex, proteasome CTL activity was highest in the cytosol and then ~50% lower in nuclear fractions; ~15-20% of total CTL activity was in pure mitochondrial fractions. With in-gel activity assay, 26S-singly and -doubly capped proteasomes were the dominant forms in the pig cerebral cortex. With a novel in situ histochemical activity assay, MG132-inhibitable proteasome CTL activity was localized to the neuropil, as a mosaic, and to cell bodies, nuclei, and centrosome-like perinuclear satellites. In piglets treated intravenously with pyrazolone derivative and chlorpromazine over 24 h, brain proteasome CTL activity was modestly increased. Discussion This study shows that the proteasome in the pig brain has relative regional uniformity, prominent nuclear and perinuclear presence with catalytic activity, a mitochondrial association with activity, 26S-single cap dominance, and indications from small molecule systemic administration of pyrazolone derivative and chlorpromazine that brain proteasome function appears safely activable.
Collapse
Affiliation(s)
- Adriana Amrein Almira
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - May W. Chen
- Departments of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Nagat El Demerdash
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Cameron Javdan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Dongseok Park
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jennifer K. Lee
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lee J. Martin
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Pathobiology Graduate Training Program, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
14
|
Cavalloro V, Marchesi N, Linciano P, Rossi D, Campagnoli LIM, Fossati A, Ahmed KM, Malacrida A, Miloso M, Mazzeo G, Abbate S, Longhi G, Ambrosio FA, Costa G, Alcaro S, Pascale A, Martino E, Collina S. Neurodegeneration: can metabolites from Eremurus persicus help? Front Pharmacol 2024; 15:1309766. [PMID: 38370479 PMCID: PMC10873958 DOI: 10.3389/fphar.2024.1309766] [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: 10/08/2023] [Accepted: 01/19/2024] [Indexed: 02/20/2024] Open
Abstract
The number of patients affected by neurodegenerative diseases is increasing worldwide, and no effective treatments have been developed yet. Although precision medicine could represent a powerful tool, it remains a challenge due to the high variability among patients. To identify molecules acting with innovative mechanisms of action, we performed a computational investigation using SAFAN technology, focusing specifically on HuD. This target belongs to the human embryonic lethal abnormal visual-like (ELAV) proteins and plays a key role in neuronal plasticity and differentiation. The results highlighted that the molecule able to bind the selected target was (R)-aloesaponol-III-8-methyl ether [(R)-ASME], a metabolite extracted from Eremurus persicus. Notably, this molecule is a TNF-α inhibitor, a cytokine involved in neuroinflammation. To obtain a suitable amount of (R)-ASME to confirm its activity on HuD, we optimized the extraction procedure. Together with ASME, another related metabolite, germichrysone, was isolated. Both ASME and germichrysone underwent biological investigation, but only ASME confirmed its ability to bind HuD. Given the multifactorial nature of neurodegenerative diseases, we decided to investigate ASME as a proteasome activator, being molecules endowed with this kind of activity potentially able to counteract aggregations of dysregulated proteins. ASME was able to activate the considered target both in enzymatic and cellular assays. Therefore, ASME may be considered a promising hit in the fight against neurodegenerative diseases.
Collapse
Affiliation(s)
- Valeria Cavalloro
- Department of Earth and Environmental Sciences, University of Pavia, Pavia, Italy
- National Biodiversity Future Center, Palermo, Italy
| | | | | | - Daniela Rossi
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | | | - Alice Fossati
- Department of Earth and Environmental Sciences, University of Pavia, Pavia, Italy
- National Biodiversity Future Center, Palermo, Italy
| | - Karzan Mahmood Ahmed
- Department of Chemistry, College of Education, University of Garmian, Kalar, Iraq
| | - Alessio Malacrida
- School of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
- Experimental Neurology Unit, University of Milano-Bicocca, Monza, Italy
| | - Mariarosaria Miloso
- School of Medicine and Surgery, University of Milan-Bicocca, Monza, Italy
- Experimental Neurology Unit, University of Milano-Bicocca, Monza, Italy
| | - Giuseppe Mazzeo
- Department of Molecular and Translational Medicine, Università di Brescia, Brescia, Italy
| | - Sergio Abbate
- Department of Molecular and Translational Medicine, Università di Brescia, Brescia, Italy
| | - Giovanna Longhi
- Department of Molecular and Translational Medicine, Università di Brescia, Brescia, Italy
| | - Francesca Alessandra Ambrosio
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro, Campus “S. Venuta”, Catanzaro, Italy
| | - Giosuè Costa
- Department of Health Sciences, Campus “S. Venuta”, “Magna Græcia” University of Catanzaro, Catanzaro, Italy
- Net4Science Academic Spin-Off, University “Magna Græcia” of Catanzaro, Campus “S. Venuta”, Catanzaro, Italy
| | - Stefano Alcaro
- Department of Health Sciences, Campus “S. Venuta”, “Magna Græcia” University of Catanzaro, Catanzaro, Italy
- Net4Science Academic Spin-Off, University “Magna Græcia” of Catanzaro, Campus “S. Venuta”, Catanzaro, Italy
- Associazione CRISEA–Centro di Ricerca e Servizi Avanzati per l’Innovazione Rurale, Italy
| | - Alessia Pascale
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Emanuela Martino
- Department of Earth and Environmental Sciences, University of Pavia, Pavia, Italy
- National Biodiversity Future Center, Palermo, Italy
| | - Simona Collina
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
15
|
Upadhyay A, Joshi V. Proteasome Activators and Ageing: Restoring Proteostasis Using Small Molecules. Subcell Biochem 2024; 107:21-41. [PMID: 39693018 DOI: 10.1007/978-3-031-66768-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Ageing is an inevitable phenomenon that remains under control of a plethora of signalling pathways and regulatory mechanisms. Slowing of cellular homeostasis and repair pathways, declining genomic and proteomic integrity, and deficient stress regulatory machinery may cause accumulating damage triggering initiation of pathways leading to ageing-associated changes. Multiple genetic studies in small laboratory organisms focused on the manipulation of proteasomal activities have shown promising results in delaying the age-related decline and improving the lifespan. In addition, a number of studies indicate a prominent role of small molecule-based proteasome activators showing positive results in ameliorating the stress conditions, protecting degenerating neurons, restoring cognitive functions, and extending life span of organisms. In this chapter, we provide a brief overview of the multi-enzyme proteasome complex, its structure, subunit composition and variety of cellular functions. We also highlight the strategies applied in the past to modulate the protein degradation efficiency of proteasome and their impact on rebalancing the proteostasis defects. Finally, we provide a descriptive account of proteasome activation mechanisms and small molecule-based strategies to improve the overall organismal health and delay the development of age-associated pathologies.
Collapse
Affiliation(s)
- Arun Upadhyay
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Department of Bioscience and Biomedical Engineering, Indian Institute of Technology Bhilai, Chhattisgarh, India.
| | - Vibhuti Joshi
- Department of Biotechnology, Bennett University, Greater Noida, Uttar Pradesh, India.
| |
Collapse
|
16
|
Moyano P, Sola E, Naval MV, Guerra-Menéndez L, Fernández MDLC, del Pino J. Neurodegenerative Proteinopathies Induced by Environmental Pollutants: Heat Shock Proteins and Proteasome as Promising Therapeutic Tools. Pharmaceutics 2023; 15:2048. [PMID: 37631262 PMCID: PMC10458078 DOI: 10.3390/pharmaceutics15082048] [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: 06/29/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
Environmental pollutants' (EPs) amount and diversity have increased in recent years due to anthropogenic activity. Several neurodegenerative diseases (NDs) are theorized to be related to EPs, as their incidence has increased in a similar way to human EPs exposure and they reproduce the main ND hallmarks. EPs induce several neurotoxic effects, including accumulation and gradual deposition of misfolded toxic proteins, producing neuronal malfunction and cell death. Cells possess different mechanisms to eliminate these toxic proteins, including heat shock proteins (HSPs) and the proteasome system. The accumulation and deleterious effects of toxic proteins are induced through HSPs and disruption of proteasome proteins' homeostatic function by exposure to EPs. A therapeutic approach has been proposed to reduce accumulation of toxic proteins through treatment with recombinant HSPs/proteasome or the use of compounds that increase their expression or activity. Our aim is to review the current literature on NDs related to EP exposure and their relationship with the disruption of the proteasome system and HSPs, as well as to discuss the toxic effects of dysfunction of HSPs and proteasome and the contradictory effects described in the literature. Lastly, we cover the therapeutic use of developed drugs and recombinant proteasome/HSPs to eliminate toxic proteins and prevent/treat EP-induced neurodegeneration.
Collapse
Affiliation(s)
- Paula Moyano
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain;
| | - Emma Sola
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain;
| | - María Victoria Naval
- Department of Pharmacology, Pharmacognosy and Bothanic, Pharmacy School, Complutense University of Madrid, 28041 Madrid, Spain
| | - Lucia Guerra-Menéndez
- Department of Physiology, Medicine School, San Pablo CEU University, 28003 Madrid, Spain
| | - Maria De la Cabeza Fernández
- Department of Chemistry and Pharmaceutical Sciences, Pharmacy School, Complutense University of Madrid, 28041 Madrid, Spain
| | - Javier del Pino
- Department of Pharmacology and Toxicology, Veterinary School, Complutense University of Madrid, 28040 Madrid, Spain;
| |
Collapse
|
17
|
Guo X, Mutch M, Torres AY, Nano M, McDonald D, Chen Z, Montell C, Dai W, Montell DJ. Rescue of proteotoxic stress and neurodegeneration by the Zn 2+ transporter ZIP7. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541645. [PMID: 37292980 PMCID: PMC10245811 DOI: 10.1101/2023.05.22.541645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Proteotoxic stress drives numerous degenerative diseases. In response to misfolded proteins, cells adapt by activating the unfolded protein response (UPR), including endoplasmic reticulum-associated protein degradation (ERAD). However persistent stress triggers apoptosis. Enhancing ERAD is a promising therapeutic approach for protein misfolding diseases. From plants to humans, loss of the Zn2+ transporter ZIP7 causes ER stress, however the mechanism is unknown. Here we show that ZIP7 enhances ERAD and that cytosolic Zn2+ is limiting for deubiquitination of client proteins by the Rpn11 Zn2+ metalloproteinase as they enter the proteasome in Drosophila and human cells. ZIP7 overexpression rescues defective vision caused by misfolded rhodopsin in Drosophila. Thus ZIP7 overexpression may prevent diseases caused by proteotoxic stress, and existing ZIP inhibitors may be effective against proteasome-dependent cancers.
Collapse
Affiliation(s)
- Xiaoran Guo
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110
- present address: Biochemistry Department, Stanford University, Stanford, CA 94305
| | - Morgan Mutch
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110
| | - Alba Yurani Torres
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110
| | - Maddalena Nano
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110
| | - Drew McDonald
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110
| | - Zijing Chen
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110
| | - Craig Montell
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110
| | - Wei Dai
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Denise J. Montell
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93110
| |
Collapse
|
18
|
Witte K, Schneider-Burrus S, Salinas G, Mössner R, Ghoreschi K, Wolk K, Sabat R. Blood T Helper Memory Cells: A Tool for Studying Skin Inflammation in HS? Int J Mol Sci 2023; 24:ijms24108854. [PMID: 37240200 DOI: 10.3390/ijms24108854] [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: 03/15/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Hidradenitis suppurativa (HS) is an inflammatory skin disease characterized by painful lesions on intertriginous body areas such as the axillary, inguinal, and perianal sites. Given the limited treatment options for HS, expanding our knowledge of its pathogenetic mechanisms is a prerequisite for novel therapeutic developments. T cells are assumed to play a crucial role in HS pathogenesis. However, it is currently unknown whether blood T cells show specific molecular alterations in HS. To address this, we studied the molecular profile of CD4+ memory T (Thmem) cells purified from the blood of patients with HS and matched healthy participants. About 2.0% and 1.9% of protein-coding transcripts were found to be up- and down-regulated in blood HS Thmem cells, respectively. These differentially expressed transcripts (DETs) are known to be involved in nucleoside triphosphate/nucleotide metabolic processes, mitochondrion organization, and oxidative phosphorylation. The detected down-regulation of transcripts involved in oxidative phosphorylation suggest a metabolic shift of HS Thmem cells towards glycolysis. The inclusion of transcriptome data from skin from HS patients and healthy participants in the analyses revealed that in HS skin lesions, the expression pattern of transcripts identified as DETs in blood HS Thmem cells was very similar to the expression pattern of the totality of protein-coding transcripts. Furthermore, there was no significant association between the extent of the expressional changes in the DETs of blood HS Thmem cells and the extent of the expressional changes in these transcripts in HS skin lesions compared to healthy donor skin. Additionally, a gene ontology enrichment analysis did not demonstrate any association of the DETs of blood HS Thmem cells with skin disorders. Instead, there were associations with different neurological diseases, non-alcoholic fatty liver disease, and thermogenesis. The levels of most DETs linked to neurological diseases showed a positive correlation to each other, suggesting common regulatory mechanisms. In summary, the transcriptomic changes in blood Thmem cells observed in patients with manifest cutaneous HS lesions do not appear to be characteristic of the molecular changes in the skin. Instead, they could be useful for studying comorbidities and identifying corresponding blood biomarkers in these patients.
Collapse
Affiliation(s)
- Katrin Witte
- Psoriasis Research and Treatment Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Interdisciplinary Group of Molecular Immunopathology, Dermatology/Medical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
| | - Sylke Schneider-Burrus
- Psoriasis Research and Treatment Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Center for Dermatosurgery, Havelklinik Berlin, 13595 Berlin, Germany
| | - Gabriela Salinas
- NGS-Integrative Genomics Core Unit, Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Rotraut Mössner
- Department of Dermatology, Georg-August-University Goettingen, 37073 Goettingen, Germany
| | - Kamran Ghoreschi
- Department of Dermatology, Venereology and Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Kerstin Wolk
- Psoriasis Research and Treatment Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Interdisciplinary Group of Molecular Immunopathology, Dermatology/Medical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
| | - Robert Sabat
- Psoriasis Research and Treatment Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
- Interdisciplinary Group of Molecular Immunopathology, Dermatology/Medical Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| |
Collapse
|
19
|
Staerz S, Lisabeth EM, Njomen E, Dexheimer TS, Neubig RR, Tepe JJ. Development of a Cell-Based AlphaLISA Assay for High-Throughput Screening for Small Molecule Proteasome Modulators. ACS OMEGA 2023; 8:15650-15659. [PMID: 37151549 PMCID: PMC10157846 DOI: 10.1021/acsomega.3c01158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/12/2023] [Indexed: 05/09/2023]
Abstract
The balance between protein degradation and protein synthesis is a highly choreographed process generally called proteostasis. Most intracellular protein degradation occurs through the ubiquitin-proteasome system (UPS). This degradation takes place through either a ubiquitin-dependent or a ubiquitin-independent proteasomal pathway. The ubiquitin-independent pathway selectively targets unfolded proteins, including intrinsically disordered proteins (IDPs). Dysregulation of proteolysis can lead to the accumulation of IDPs, seen in the pathogenesis of various diseases, including cancer and neurodegeneration. Therefore, the enhancement of the proteolytic activity of the 20S proteasome using small molecules has been identified as a promising pathway to combat IDP accumulation. Currently, there are a limited number of known small molecules that enhance the activity of the 20S proteasome, and few are observed to exhibit enhanced proteasome activity in cell culture. Herein, we describe the development of a high-throughput screening assay to identify cell-permeable proteasome enhancers by utilizing an AlphaLISA platform that measures the degradation of a GFP conjugated intrinsically disordered protein, ornithine decarboxylase (ODC). Through the screening of the Prestwick and NIH Clinical Libraries, a kinase inhibitor, erlotinib, was identified as a new 20S proteasome enhancer, which enhances the degradation of ODC in cells and α-synuclein in vitro.
Collapse
Affiliation(s)
- Sophia
D. Staerz
- Department
of Chemistry, Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Erika M. Lisabeth
- Department
of Chemistry, Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Evert Njomen
- Department
of Chemistry, Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Thomas S. Dexheimer
- Department
of Chemistry, Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Richard R. Neubig
- Department
of Chemistry, Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Jetze J. Tepe
- Department
of Chemistry, Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
20
|
Kurata K, Samur MK, Liow P, Wen K, Yamamoto L, Liu J, Morelli E, Gulla A, Tai YT, Qi J, Hideshima T, Anderson KC. BRD9 Degradation Disrupts Ribosome Biogenesis in Multiple Myeloma. Clin Cancer Res 2023; 29:1807-1821. [PMID: 36780189 PMCID: PMC10150249 DOI: 10.1158/1078-0432.ccr-22-3668] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/12/2023] [Accepted: 02/09/2023] [Indexed: 02/14/2023]
Abstract
PURPOSE BRD9 is a defining component of the noncanonical SWI/SNF complex, which regulates gene expression by controlling chromatin dynamics. Although recent studies have found an oncogenic role for BRD9 in multiple cancer types including multiple myeloma, its clinical significance and oncogenic mechanism have not yet been elucidated. Here, we sought to identify the clinical and biological impact of BRD9 in multiple myeloma, which may contribute to the development of novel therapeutic strategies. EXPERIMENTAL DESIGN We performed integrated analyses of BRD9 in vitro and in vivo using multiple myeloma cell lines and primary multiple myeloma cells in established preclinical models, which identified the molecular functions of BRD9 contributing to multiple myeloma cell survival. RESULTS We found that high BRD9 expression was a poor prognostic factor in multiple myeloma. Depleting BRD9 by genetic (shRNA) and pharmacologic (dBRD9-A; proteolysis-targeting chimera; BRD9 degrader) approaches downregulated ribosome biogenesis genes, decreased the expression of the master regulator MYC, and disrupted the protein-synthesis maintenance machinery, thereby inhibiting multiple myeloma cell growth in vitro and in vivo in preclinical models. Importantly, we identified that the expression of ribosome biogenesis genes was associated with the disease progression and prognosis of patients with multiple myeloma. Our results suggest that BRD9 promotes gene expression by predominantly occupying the promoter regions of ribosome biogenesis genes and cooperating with BRD4 to enhance the transcriptional function of MYC. CONCLUSIONS Our study identifies and validates BRD9 as a novel therapeutic target in preclinical models of multiple myeloma, which provides the framework for the clinical evaluation of BRD9 degraders to improve patient outcome.
Collapse
Affiliation(s)
- Keiji Kurata
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Mehmet K. Samur
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute and Harvard School of Public Health, Boston, Massachusetts
| | - Priscilla Liow
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kenneth Wen
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Leona Yamamoto
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Jiye Liu
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Eugenio Morelli
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Annamaria Gulla
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | - Yu-Tzu Tai
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Jun Qi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Teru Hideshima
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kenneth C. Anderson
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
21
|
Vasilopoulou MA, Gioran A, Theodoropoulou M, Koutsaviti A, Roussis V, Ioannou E, Chondrogianni N. Healthspan improvement and anti-aggregation effects induced by a marine-derived structural proteasome activator. Redox Biol 2022; 56:102462. [PMID: 36095970 PMCID: PMC9482115 DOI: 10.1016/j.redox.2022.102462] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/28/2022] [Accepted: 08/28/2022] [Indexed: 11/15/2022] Open
Abstract
Proteasome activation has been shown to promote cellular and organismal healthspan and to protect against aggregation-related conditions, such as Alzheimer's disease (AD). Various natural compounds have been described for their proteasome activating properties but scarce data exist on marine metabolites that often possess unique chemical structures, exhibiting pronounced bioactivities with novel mechanisms of action. In this study, we have identified for the first time a marine structural proteasome activator, namely (1R,3E,6R,7Z,11S,12S)-dolabella-3,7,18-trien-6,17-olide (DBTO). DBTO activates the 20S proteasome complex in cell-free assays but also in cellulo. Continuous supplementation of human primary fibroblasts with DBTO throughout their cellular lifespan confers an improved healthspan while ameliorated health status is also observed in wild type (wt) Caenorhabditis elegans (C. elegans) nematodes supplemented with DBTO. Furthermore, treatment of various AD nematode models, as well as of human cells of neuronal origin challenged with exogenously added Aβ peptide, with DBTO results in enhanced protection against Aβ-induced proteotoxicity. In total, our results reveal the first structural proteasome activator derived from the marine ecosystem and highlight its potential as a compound that might be used for healthspan maintenance and preventive strategies against proteinopathies, such as AD. (1R,3E,6R,7Z,11S,12S)-dolabella-3,7,18-trien-6,17-olide (DBTO) is a structural proteasome activator. DBTO is the first identified marine structural proteasome activator. DBTO positively modulates cellular healthspan and organismal health status. DBTO confers protection against Aβ-induced proteotoxicity.
Collapse
|
22
|
Cirqueira L, Stock L, Treptow W. Concentration-Dependent Thermodynamic Analysis of the Partition Process of Small Ligands into Proteins. Comput Struct Biotechnol J 2022; 20:4885-4891. [PMID: 36147679 PMCID: PMC9468351 DOI: 10.1016/j.csbj.2022.08.049] [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: 06/02/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 11/28/2022] Open
Abstract
In the category of functional low-affinity interactions, small ligands may interact with multiple protein sites in a highly degenerate manner. Better conceived as a partition phenomenon at the molecular interface of proteins, such low-affinity interactions appear to be hidden to our current experimental resolution making their structural and functional characterization difficult in the low concentration regime of physiological processes. Characterization of the partition phenomenon under higher chemical forces could be a relevant strategy to tackle the problem provided the results can be scaled back to the low concentration range. Far from being trivial, such scaling demands a concentration-dependent understanding of self-interactions of the ligands, structural perturbations of the protein, among other molecular effects. Accordingly, we elaborate a novel and detailed concentration-dependent thermodynamic analysis of the partition process of small ligands aiming at characterizing the stability and structure of the dilute phenomenon from high concentrations. In analogy to an “aggregate” binding constant of a small molecule over multiple sites of a protein receptor, the model defines the stability of the process as a macroscopic equilibrium constant for the partition number of ligands that can be used to analyze biochemical and functional data of two-component systems driven by low-affinity interactions. Acquisition of such modeling-based structural information is expected to be highly welcome by revealing more traceable protein-binding spots for non-specific ligands.
Collapse
|
23
|
Apoptosis and (in) Pain—Potential Clinical Implications. Biomedicines 2022; 10:biomedicines10061255. [PMID: 35740277 PMCID: PMC9219669 DOI: 10.3390/biomedicines10061255] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 02/06/2023] Open
Abstract
The deregulation of apoptosis is involved in the development of several pathologies, and recent evidence suggests that apoptosis may be involved in chronic pain, namely in neuropathic pain. Neuropathic pain is a chronic pain state caused by primary damage or dysfunction of the nervous system; however, the details of the molecular mechanisms have not yet been fully elucidated. Recently, it was found that nerve endings contain transient receptor potential (TRP) channels that sense and detect signals released by injured tissues and respond to these damage signals. TRP channels are similar to the voltage-gated potassium channels or nucleotide-gated channels that participate in calcium and magnesium homeostasis. TRP channels allowing calcium to penetrate into nerve terminals can activate apoptosis, leading to nerve terminal destruction. Further, some TRPs are activated by acid and reactive oxygen species (ROS). ROS are mainly produced in the mitochondrial respiratory chain, and an increase in ROS production and/or a decrease in the antioxidant network may induce oxidative stress (OS). Depending on the OS levels, they can promote cellular proliferation and/or cell degeneration or death. Previous studies have indicated that proinflammatory cytokines, such as tumor necrosis factor-α (TNF-α), play an important role in the peripheral mediation of neuropathic pain. This article aims to perform a review of the involvement of apoptosis in pain, particularly the role of OS and neuroinflammation, and the clinical relevance of this knowledge. The potential discovery of new biomarkers and therapeutic targets can result in the development of more effective and targeted drugs to treat chronic pain, namely neuropathic pain. Highlights: Oxidative stress and neuroinflammation can activate cell signaling pathways that can lead to nerve terminal destruction by apoptosis. These could constitute potential new pain biomarkers and targets for therapy in neuropathic pain.
Collapse
|