1
|
Stanciu SM, Jurcut R, Dragoi Galrinho R, Stefani C, Miricescu D, Rusu IR, Prisacariu GS, Mititelu R. From Molecular to Radionuclide and Pharmacological Aspects in Transthyretin Cardiac Amyloidosis. Int J Mol Sci 2024; 26:146. [PMID: 39796004 PMCID: PMC11719977 DOI: 10.3390/ijms26010146] [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: 11/24/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Amyloidosis is a rare pathology characterized by protein deposits in various organs and tissues. Cardiac amyloidosis (CA) can be caused by various protein deposits, but transthyretin amyloidosis (ATTR) and immunoglobulin light chain (AL) are the most frequent pathologies. Protein misfolding can be induced by several factors such as oxidative stress, genetic mutations, aging, chronic inflammation, and neoplastic disorders. In ATTR cardiomyopathy (ATTR-CM), the amyloid fibrils can be found in the myocardium interstitial space and are associated with arrhythmias and heart failure. In pathological situations, the transthyretin (TTR) configuration is destroyed by proteolytic action, leading to monomers that further misfold and aggregate to form the amyloid fibrils. 99mTc-Pyrophosphate (99m-Tc-PYP), 99mTc 3,3-diphosphono-1,2-propanodicarboxylic acid (99m-Tc-DPD) and 99m-Tc hydroxy-methylene-Dyphosphonate (99m-Tc-HMDP) are used to detect myocardium amyloid deposits due to their ability to detect calcium ions that are present in the amyloid fibrils through dystrophic calcification. ATTR-CM therapy acts on different stages of the amyloidogenic process, including liver TTR synthesis, TTR tetramer destabilization, and misfolding of the monomers. The main aim of this narrative review is to present ATTR-CM, starting with molecular changes regarding the protein misfolding process and radionuclide aspects and finishing with pharmacological approaches.
Collapse
Affiliation(s)
- Silviu Marcel Stanciu
- Department of Internal Medicine and Gastroenterology, Carol Davila University of Medicine and Pharmacy, Central Military Emergency University Hospital, 010825 Bucharest, Romania;
| | - Ruxandra Jurcut
- Department of Cardiology, Carol Davila University of Medicine and Pharmacy, Institute of Cardiovascular Diseases “Prof CC Iliescu”, 022322 Bucharest, Romania;
| | - Ruxandra Dragoi Galrinho
- Department of Cardiology and Cardiovascular Surgery, University and Emergency Hospital, 050098 Bucharest, Romania
| | - Constantin Stefani
- Department I of Family Medicine and Clinical Base, “Dr. Carol Davila” Central Military Emergency University Hospital, 010825 Bucharest, Romania;
| | - Daniela Miricescu
- Discipline of Biochemistry, Faculty of Dentistry, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Ioana Ruxandra Rusu
- Discipline of Anatomy, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Georgiana Sabina Prisacariu
- Clinic of Nuclear Medicine Central University Emergency Military Hospital “Dr Carol Davila”, 10825 Bucharest, Romania; (G.S.P.); (R.M.)
| | - Raluca Mititelu
- Clinic of Nuclear Medicine Central University Emergency Military Hospital “Dr Carol Davila”, 10825 Bucharest, Romania; (G.S.P.); (R.M.)
- Department of Nuclear Medicine, University of Medicine and Pharmacy Carol Davila, 030147 Bucharest, Romania
| |
Collapse
|
2
|
Jing J, Wang J, Xiang X, Yin S, Tang J, Wang L, Jia G, Liu G, Chen X, Tian G, Cai J, Kang B, Che L, Zhao H. Selenomethionine alleviates chronic heat stress-induced breast muscle injury and poor meat quality in broilers via relieving mitochondrial dysfunction and endoplasmic reticulum stress. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 16:363-375. [PMID: 38362514 PMCID: PMC10867585 DOI: 10.1016/j.aninu.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 12/18/2023] [Accepted: 12/25/2023] [Indexed: 02/17/2024]
Abstract
In the present study, the chronic heat stress (CHS) broiler model was developed to investigate the potential protection mechanism of organic selenium (selenomethionine, SeMet) on CHS-induced skeletal muscle growth retardation and poor meat quality. Four hundred Arbor Acres male broilers (680 ± 70 g, 21 d old) were grouped into 5 treatments with 8 replicates of 10 broilers per replicate. Broilers in the control group were raised in a thermoneutral environment (22 ± 2 °C) and fed with a basal diet. The other four treatments were exposed to hyperthermic conditions (33 ± 2 °C, 24 h in each day) and fed on the basal diet supplied with SeMet at 0.0, 0.2, 0.4, and 0.6 mg Se/kg, respectively, for 21 d. Results showed that CHS reduced (P < 0.05) the growth performance, decreased (P < 0.05) the breast muscle weight and impaired the meat quality of breast muscle in broilers. CHS induced protein metabolic disorder in breast muscle, which increased (P < 0.05) the expression of caspase 3, caspase 8, caspase 9 and ubiquitin proteasome system related genes, while decreased the protein expression of P-4EBP1. CHS also decreased the antioxidant capacity and induced mitochondrial stress and endoplasmic reticulum (ER) stress in breast muscle, which increased (P < 0.05) the ROS levels, decreased the concentration of ATP, increased the protein expression of HSP60 and CLPX, and increased (P < 0.05) the expression of ER stress biomarkers. Dietary SeMet supplementation linearly increased (P < 0.05) breast muscle Se concentration and exhibited protective effects via up-regulating the expression of the selenotranscriptome and several key selenoproteins, which increased (P < 0.05) body weight, improved meat quality, enhanced antioxidant capacity and mitigated mitochondrial stress and ER stress. What's more, SeMet suppressed protein degradation and improved protein biosynthesis though inhibiting the caspase and ubiquitin proteasome system and promoting the mTOR-4EBP1 pathway. In conclusion, dietary SeMet supplementation increases the expression of several key selenoproteins, alleviates mitochondrial dysfunction and ER stress, improves protein biosynthesis, suppresses protein degradation, thus increases the body weight and improves meat quality of broilers exposed to CHS.
Collapse
Affiliation(s)
- Jinzhong Jing
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Jiayi Wang
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Xiaoyu Xiang
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Shenggang Yin
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Jiayong Tang
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Longqiong Wang
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Gang Jia
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Guangmang Liu
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Xiaoling Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Gang Tian
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Jingyi Cai
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Bo Kang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Lianqiang Che
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Hua Zhao
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, of China Ministry of Agriculture and Rural Affairs, of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| |
Collapse
|
3
|
Zhou L, Jeong IH, Xue S, Xue M, Wang L, Li S, Liu R, Jeong GH, Wang X, Cai J, Yin J, Huang B. Inhibition of the Ubiquitin Transfer Cascade by a Peptidomimetic Foldamer Mimicking the E2 N-Terminal Helix. J Med Chem 2023; 66:491-502. [PMID: 36571278 DOI: 10.1021/acs.jmedchem.2c01459] [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: 12/27/2022]
Abstract
The enzymatic cascades for ubiquitin transfer regulate key cellular processes and are the intense focus of drug development for treating cancer and neurodegenerative diseases. E1 is at the apex of the UB transfer cascade, and molecules inhibiting E1 have shown promising activities against cancer cell proliferation. Compared to small molecules, peptidomimetics have emerged as powerful tools to disrupt the protein-protein interactions (PPI) with less drug resistance and high stability in the cell. Herein, we harnessed the D-sulfono-γ-AA peptide to mimic the N-terminal helix of E2 and thereby inhibit E1-E2 interaction. Two stapled peptidomimetics, M1-S1 and M1-S2, were identified as effective inhibitors to block UB transfer from E1 to E2, as shown by in vitro and cellular assays. Our work suggested that PPIs with the N-terminal helix of E2 at the E1-E2 and E2-E3 interfaces could be a promising target for designing inhibitors against protein ubiquitination pathways in the cell.
Collapse
Affiliation(s)
- Li Zhou
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, Georgia30303, United States
| | - In Ho Jeong
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, Georgia30303, United States
| | - Songyi Xue
- Department of Chemistry, University of South Florida, Tampa, Florida33620, United States
| | - Menglin Xue
- Department of Chemistry, University of South Florida, Tampa, Florida33620, United States
| | - Lei Wang
- Department of Chemistry, University of South Florida, Tampa, Florida33620, United States
| | - Sihao Li
- Department of Chemistry, University of South Florida, Tampa, Florida33620, United States
| | - Ruochuan Liu
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, Georgia30303, United States
| | - Geon Ho Jeong
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, Georgia30303, United States
| | - Xiaoyu Wang
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, Georgia30303, United States
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, Florida33620, United States
| | - Jun Yin
- Department of Chemistry and Center for Diagnostics & Therapeutics, Georgia State University, Atlanta, Georgia30303, United States
| | - Bo Huang
- Department of Chemistry, University of South Florida, Tampa, Florida33620, United States
| |
Collapse
|
4
|
Wan S, Cao J, Chen S, Yang J, Wang H, Wang C, Li K, Yang L. Construction of noninvasive prognostic model of bladder cancer patients based on urine proteomics and screening of natural compounds. J Cancer Res Clin Oncol 2023; 149:281-296. [PMID: 36562811 DOI: 10.1007/s00432-022-04524-x] [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: 10/16/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Bladder cancer (BCa) has a high incidence and recurrence rate worldwide. So far, there is no noninvasive detection of BCa therapy and prognosis based on urine multi-omics. Therefore, it is necessary to explore noninvasive predictive models and novel treatment modalities for BCa. METHODS First, we performed protein analysis of urine from five BCa patients and five healthy individuals using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Combining multi-omics data to mine particular and sensitive molecules to predict BCa prognosis. Second, urine proteomics data were combined with TCGA transcriptome data to select differential genes that were specifically highly expressed in urine and tissues. Further, the Lasso equation was used to screen specific molecules to construct a noninvasive prediction model of BCa. Finally, natural compounds of specific molecules were selected by combined network pharmacology and molecular docking to complete molecular structure docking. RESULTS A noninvasive predictive model was constructed using PSMB5, P4HB, S100A16, GET3, CNP, TFRC, DCXR, and MPZL1, specific molecules screened by multi-omics, and clinical features, which had good predictive value at 1, 3, and 5 years of prediction. High expression of these target genes suggests a poor prognosis in patients with BCa, and they were mainly involved in cell adhesion molecules and the IGF pathway. In addition, the corresponding drugs and natural compounds were selected by network pharmacology, and the molecular structure 7NHT of PSMB5 was found to be well docked to Ellagic acid, a natural compound in Hetaoren that we found. The 3D structure 6I7S of P4HB was able to bind to Stigmasterol in Shanzha stably, and the structure 6WRV of TFRC as an iron transport carrier was also able to bind to Stigmasterol in Shanzha stably. The structures 1WOJ, 3D3W, and 6IGW of CNP, DCXR, and MPZL1 can also play an important role in combination with the natural compounds (S)-Stylopine, Kryptoxanthin, and Sitosterol in Maqianzi, Yumixu, and Laoguancao. CONCLUSION The noninvasive prediction model based on urinomics had excellent potential in predicting the prognosis of patients with BCa. The multi-omics screening of specific molecules combined with pharmacology and compound molecular docking can promote the research and development of novel drugs.
Collapse
Affiliation(s)
- Shun Wan
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730000, China.,Gansu Province Clinical Research Center for Urology, Lanzhou, 730000, China
| | - Jinlong Cao
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730000, China.,Gansu Province Clinical Research Center for Urology, Lanzhou, 730000, China
| | - Siyu Chen
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730000, China.,Gansu Province Clinical Research Center for Urology, Lanzhou, 730000, China
| | - Jianwei Yang
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Huabin Wang
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730000, China.,Gansu Province Clinical Research Center for Urology, Lanzhou, 730000, China
| | - Chenyang Wang
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730000, China.,Gansu Province Clinical Research Center for Urology, Lanzhou, 730000, China
| | - Kunpeng Li
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730000, China.,Gansu Province Clinical Research Center for Urology, Lanzhou, 730000, China
| | - Li Yang
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, 730000, China. .,Gansu Province Clinical Research Center for Urology, Lanzhou, 730000, China.
| |
Collapse
|
5
|
Liu J, Mi J, Liu S, Chen H, Jiang L. PSMB5 overexpression is correlated with tumor proliferation and poor prognosis in hepatocellular carcinoma. FEBS Open Bio 2022; 12:2025-2041. [PMID: 36062301 PMCID: PMC9623531 DOI: 10.1002/2211-5463.13479] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 07/14/2022] [Accepted: 08/11/2022] [Indexed: 01/25/2023] Open
Abstract
Aberrant expression of members of the proteasome subunit beta (PSMB) family (including PSMB2, PSMB4, PSMB7 and PSMB8) has been reported in hepatocellular carcinoma (HCC). However the role of PSMB5 in HCC is unclear. To address this issue, we examined the expression of PSMB5 in HCC tissues using the The Cancer Genome Atlas, International Cancer Genome Consortium and Gene Expression Omnibus databases. A quantitative real-time PCR and immunohistochemistry were performed to validate the expression of PSMB5 in HCC. The survival mutation status and immune cell infiltration of PSMB5 were also evaluated in HCC. We then examined the effect of knocking down PSMB5 expression through RNA interference in the HCC cell line Huh7. High expression of PSMB5 was observed in HCC tissues and was associated with poor prognosis. PSMB5 expression and clinical characteristics were then incorporated to build a prognostic nomogram. We observed that PSMB5 expression was closely related to the abundance of B cells, CD4+ T cells, CD8+ T cells, dendritic cell macrophages and neutrophils. Moreover silencing of PSMB5 in Huh7 significantly suppressed cell proliferation and migration at the same time as increasing apoptosis. Inhibition of the phosphatidylinositol-3-kinase/Akt/mechanistic target of rapamycin pathway was observed after PSMB5 downregulation in Huh7 cells. Our findings suggest that PSMB5 may promote the proliferation of HCC cells by inactivating the phosphatidylinositol-3-kinase/Akt/mechanistic target of rapamycin signaling pathway and thus PSMB5 may have potential as a biomarker for diagnosis and prognosis of HCC.
Collapse
Affiliation(s)
- Jun Liu
- Guangxi Medical UniversityNanningChina
| | - Jinglin Mi
- Department of Radiation OncologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | | | | | - Li Jiang
- Department of Radiation OncologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| |
Collapse
|
6
|
Modulation of the 20S Proteasome Activity by Porphyrin Derivatives Is Steered through Their Charge Distribution. Biomolecules 2022; 12:biom12060741. [PMID: 35740865 PMCID: PMC9220251 DOI: 10.3390/biom12060741] [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: 04/21/2022] [Revised: 05/15/2022] [Accepted: 05/20/2022] [Indexed: 11/20/2022] Open
Abstract
Cationic porphyrins exhibit an amazing variety of binding modes and inhibition mechanisms of 20S proteasome. Depending on the spatial distribution of their electrostatic charges, they can occupy different sites on α rings of 20S proteasome by exploiting the structural code responsible for the interaction with regulatory proteins. Indeed, they can act as competitive or allosteric inhibitors by binding at the substrate gate or at the grooves between the α subunits, respectively. Moreover, the substitution of a charged moiety in the peripheral arm with a hydrophobic moiety revealed a “new” 20S functional state with higher substrate affinity and catalytic efficiency. In the present study, we expand our structure–activity relationship (SAR) analysis in order to further explore the potential of this versatile class of 20S modulators. Therefore, we have extended the study to additional macrocyclic compounds, displaying different structural features, comparing their interaction behavior on the 20S proteasome with previously investigated compounds. In particular, in order to evaluate how the introduction of a peptidic chain can affect the affinity and the interacting mechanism of porphyrins, we investigate the MTPyApi, a porphyrin derivatized with an Arg–Pro-rich antimicrobial peptide. Moreover, to unveil the role played by the porphyrin core, this was replaced with a corrole scaffold, a “contracted” version of the tetrapyrrolic ring due to the lack of a methine bridge. The analysis has been undertaken by means of integrated kinetic, Nuclear Magnetic Resonance, and computational studies. Finally, in order to assess a potential pharmacological significance of this type of investigation, a preliminary attempt has been performed to evaluate the biological effect of these molecules on MCF7 breast cancer cells in dark conditions, envisaging that porphyrins may indeed represent a powerful tool for the modulation of cellular proteostasis.
Collapse
|
7
|
Li C, Li J, Tao H, Shan J, Liu F, Deng X, Lin Y, Lin X, Fu L, Wang B, Bi Y. Differential hippocampal protein expression between normal mice and mice with the perioperative neurocognitive disorder: a proteomic analysis. Eur J Med Res 2021; 26:130. [PMID: 34732255 PMCID: PMC8565051 DOI: 10.1186/s40001-021-00599-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 10/06/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES To compare differential expression protein in hippocampal tissues from mice of perioperative neurocognitive disorder (PND) and normal control mice and to explore the possible mechanism of PND. METHODS Mice were randomly divided into a PND group (n = 9) and a control group (n = 9).The mice in the PND group were treated with open tibial fracture with intramedullary fixation under isoflurane anesthesia, while the mice in the control group received pure oxygen without surgery. The cognitive functions of the two groups were examined using Morris water maze experiment, Open field test and Fear conditioning test. The protein expression of the hippocampus of mice was analyzed by high-performance liquid chromatography-mass spectrometry (HPLC-MS/MS). Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed to explore the principal functions of dysregulated proteins. RESULTS A total of 21 proteins were differentially expressed between PND and control mice on days 1, 3, and 7 after the operation. These proteins were involved in many pathological processes, such as neuroinflammatory responses, mitochondrial oxidative stress, impaired synaptic plasticity, and neuronal cell apoptosis. Also, the dysregulated proteins were involved in MAPK, AMPK, and ErbB signaling pathways. CONCLUSION The occurrence of PND could be attributed to multiple mechanisms.
Collapse
Affiliation(s)
- Chuan Li
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, 5 Donghai Middle Road, Qingdao, Shandong, 266071, People's Republic of China
| | - Jingzhu Li
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, 5 Donghai Middle Road, Qingdao, Shandong, 266071, People's Republic of China
| | - He Tao
- Department of Anesthesiology, Dalian Medical University, Dalian, Liaoning, China
| | - Jinghua Shan
- Department of Anesthesiology, Weifang Medical University, Weifang, Shandong, China
| | - Fanghao Liu
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, 5 Donghai Middle Road, Qingdao, Shandong, 266071, People's Republic of China
| | - Xiyuan Deng
- Department of Anesthesiology, Dalian Medical University, Dalian, Liaoning, China
| | - Yanan Lin
- Department of Anesthesiology, Weifang Medical University, Weifang, Shandong, China
| | - Xu Lin
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, 5 Donghai Middle Road, Qingdao, Shandong, 266071, People's Republic of China
| | - Li Fu
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, 5 Donghai Middle Road, Qingdao, Shandong, 266071, People's Republic of China
| | - Bin Wang
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, 5 Donghai Middle Road, Qingdao, Shandong, 266071, People's Republic of China
| | - Yanlin Bi
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, 5 Donghai Middle Road, Qingdao, Shandong, 266071, People's Republic of China.
| |
Collapse
|
8
|
Ma L, Li X, Zhao X, Sun H, Kong F, Li Y, Sui Y, Xu F. Oxaliplatin promotes siMAD2L2‑induced apoptosis in colon cancer cells. Mol Med Rep 2021; 24:629. [PMID: 34278473 PMCID: PMC8281267 DOI: 10.3892/mmr.2021.12268] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 05/26/2021] [Indexed: 02/07/2023] Open
Abstract
The clinical efficacy of colorectal tumor treatment is restricted due to platinum agent resistance. Translesion DNA synthesis (TLS) has been shown to contribute to this resistance; however, the exact molecular mechanism remains unknown. The present study aimed to investigate the possible function of the core of the TLS polymerase mitotic arrest deficient 2 like 2 (MAD2L2) in drug sensitivity, in order to provide a treatment rationale for platinum‑based chemotherapy in colon cancer. In the present study, MAD2L2 was knocked down using MAD2L2‑specific small interfering (si)RNA. HCT116 and SW620 cells were treated with oxaliplatin and MG132; oxaliplatin is a platinum compound that induces DNA damage and MG132 is a potent proteasome inhibitor. Cell viability was determined using an MTT assay. Cell apoptosis was examined via flow cytometry and TUNEL assay. The activity of proteasome 26S subunit, non‑ATPase 13 (PSMD13) was detected using ELISA, while the expression levels of apoptotic‑related proteins were detected via western blotting. The results demonstrated that cells treated with oxaliplatin or MG132 alone had decreased viability, but a synergistic effect was not observed after co‑treatment. In addition, the knockdown of MAD2L2 caused by siMAD2L2 or oxaliplatin treatment increased the expression levels of the pro‑apoptotic proteins Bax and Bak and decreased the expression levels of the anti‑apoptotic protein Bcl‑2, compared with the negative control group. Moreover, MG132 alleviated the decrease in MAD2L2 expression, while reducing siMAD2L2‑induced cell apoptosis. These results indicate that oxaliplatin promotes siMAD2L2‑induced apoptosis in colon cancer cells. This process was associated with the Bcl‑2 and ubiquitin‑proteasome pathway. Overall, the present study provides a theoretical basis for improving the clinical efficacy of colon cancer by combining chemotherapy and gene therapy.
Collapse
Affiliation(s)
- Lu Ma
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
- Key Laboratory of Reproduction and Genetics, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Xin Li
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
- Key Laboratory of Reproduction and Genetics, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Xiaopeng Zhao
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
- Key Laboratory of Reproduction and Genetics, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Haotong Sun
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
- Key Laboratory of Reproduction and Genetics, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Feifei Kong
- Department of Oncology, Qufu People's Hospital, Qufu, Shandong 273100, P.R. China
| | - Yuanjie Li
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
- Key Laboratory of Reproduction and Genetics, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yu Sui
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
- Key Laboratory of Reproduction and Genetics, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Fang Xu
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
- Key Laboratory of Reproduction and Genetics, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
9
|
Abstract
Targeted protein degradation is a broad and expanding field aimed at the modulation of protein homeostasis. A focus of this field has been directed toward molecules that hijack the ubiquitin proteasome system with heterobifunctional ligands that recruit a target protein to an E3 ligase to facilitate polyubiquitination and subsequent degradation by the 26S proteasome. Despite the success of these chimeras toward a number of clinically relevant targets, the ultimate breadth and scope of this approach remains uncertain. Here we highlight recent advances in assays and tools available to evaluate targeted protein degradation, including and beyond the study of E3-targeted chimeric ligands. We note several challenges associated with degrader development and discuss various approaches to expanding the protein homeostasis toolbox.
Collapse
|
10
|
Musaus M, Navabpour S, Jarome TJ. The diversity of linkage-specific polyubiquitin chains and their role in synaptic plasticity and memory formation. Neurobiol Learn Mem 2020; 174:107286. [PMID: 32745599 DOI: 10.1016/j.nlm.2020.107286] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/15/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022]
Abstract
Over the last 20 years, a number of studies have provided strong support for protein degradation mediated by the ubiquitin-proteasome system in synaptic plasticity and memory formation. In this system, target substrates become covalently modified by the small protein ubiquitin through a series of enzymatic reactions involving hundreds of different ligases. While some substrates will acquire only a single ubiquitin, most will be marked by multiple ubiquitin modifications, which link together at specific lysine sites or the N-terminal methionine on the previous ubiquitin to form a polyubiquitin chain. There are at least eight known linkage-specific polyubiquitin chains a target protein can acquire, many of which are independent of the proteasome, and these chains can be homogenous, mixed, or branched in nature, all of which result in different functional outcomes and fates for the target substrate. However, as the focus has remained on protein degradation, much remains unknown about the role of these diverse ubiquitin chains in the brain, particularly during activity- and learning-dependent synaptic plasticity. Here, we review the different types and functions of ubiquitin chains and summarize evidence suggesting a role for these diverse ubiquitin modifications in synaptic plasticity and memory formation. We conclude by discussing how technological limitations have limited our ability to identify and elucidate the role of different ubiquitin chains in the brain and speculate on the future directions and implications of understanding linkage-specific ubiquitin modifications in activity- and learning-dependent synaptic plasticity.
Collapse
Affiliation(s)
- Madeline Musaus
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Shaghayegh Navabpour
- Fralin Biomedical Research Institute, Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, USA
| | - Timothy J Jarome
- School of Neuroscience, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA; Fralin Biomedical Research Institute, Translational Biology, Medicine and Health, Virginia Polytechnic Institute and State University, Roanoke, VA, USA; Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA.
| |
Collapse
|
11
|
Lv Y, Hu Q, Shi M, Wang W, Zheng Y, Yang Z, Peng L, Bi D, Zhang A, Hu Y. The role of PSMB5 in sodium arsenite-induced oxidative stress in L-02 cells. Cell Stress Chaperones 2020; 25:533-540. [PMID: 32301004 PMCID: PMC7192974 DOI: 10.1007/s12192-020-01104-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/16/2020] [Accepted: 03/19/2020] [Indexed: 12/26/2022] Open
Abstract
Endemic arsenism is widely distributed in the world, which can damage multiple organs, especially in skin and liver. The etiology is clear, but the mechanisms involved remain unknown. Ubiquitin-proteasome pathway (UPP) is the main pathway regulating protein degradation of which proteasome subunit beta type-5(PSMB5) plays a dominant role. This paper aims to study the role and mechanism of PSMB5 in sodium arsenite (NaAsO2)-induced oxidative stress liver injury in L-02 cells. Firstly, L-02 cells were exposed to different concentrations of NaAsO2 to establish a liver injury model of oxidative stress, and then mechanisms of oxidative stress were studied with carbobenzoxyl-leucyl-leucl-leucll-line (MG132) and knockdown PSMB5 (PSMB5-siRNA). The oxidative stress indicators, levels of 20S proteasome, the transcription and protein expression levels of PSMB5, Cu-Zn superoxide dismutase (SOD1), and glutathione peroxidase 1 (GPx1) were detected. The results demonstrated that NaAsO2 could induce oxidative stress-induced liver injury and the activity of 20S proteasome and the protein expression of PSMB5, SOD1, and GPx1 decreased. After MG132 or PSMB5-siRNA pretreatment, the gene expression of PSMB decreased. After MG132 or PSMB5-siRNA pretreatment, and then L-02 cells were treated with NaAsO2, the gene expression of PSMB remarkably decreased; however, the protein expression of SOD1 and GPx1 increased. Overall, NaAsO2 exposure could induce oxidative stress liver injury and low expression of PSMB5 in L-02 cells, and PSMB5 might play an important role in the regulation of oxidative stress by regulating the expression of SOD1 and Gpx1.
Collapse
Affiliation(s)
- Ying Lv
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Qian Hu
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Mingyang Shi
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Wen Wang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Yuancui Zheng
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Zhong Yang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Liuyu Peng
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Dingnian Bi
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Aihua Zhang
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Yong Hu
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025, Guizhou, China.
| |
Collapse
|
12
|
Osmulski PA, Karpowicz P, Jankowska E, Bohmann J, Pickering AM, Gaczyńska M. New Peptide-Based Pharmacophore Activates 20S Proteasome. Molecules 2020; 25:E1439. [PMID: 32235805 PMCID: PMC7145288 DOI: 10.3390/molecules25061439] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/13/2020] [Accepted: 03/18/2020] [Indexed: 02/01/2023] Open
Abstract
The proteasome is a pivotal element of controlled proteolysis, responsible for the catabolic arm of proteostasis. By inducing apoptosis, small molecule inhibitors of proteasome peptidolytic activities are successfully utilized in treatment of blood cancers. However, the clinical potential of proteasome activation remains relatively unexplored. In this work, we introduce short TAT peptides derived from HIV-1 Tat protein and modified with synthetic turn-stabilizing residues as proteasome agonists. Molecular docking and biochemical studies point to the α1/α2 pocket of the core proteasome α ring as the binding site of TAT peptides. We postulate that the TATs' pharmacophore consists of an N-terminal basic pocket-docking "activation anchor" connected via a β turn inducer to a C-terminal "specificity clamp" that binds on the proteasome α surface. By allosteric effects-including destabilization of the proteasomal gate-the compounds substantially augment activity of the core proteasome in vitro. Significantly, this activation is preserved in the lysates of cultured cells treated with the compounds. We propose that the proteasome-stimulating TAT pharmacophore provides an attractive lead for future clinical use.
Collapse
Affiliation(s)
- Paweł A. Osmulski
- Department of Molecular Medicine, UT Health San Antonio, Texas, TX 78245, USA;
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, Texas, TX 78245, USA
| | - Przemysław Karpowicz
- Department of Organic Chemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland;
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland;
| | - Elżbieta Jankowska
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, 80-308 Gdansk, Poland;
| | - Jonathan Bohmann
- Southwest Research Institute, San Antonio, Texas, TX 78238, USA;
| | - Andrew M. Pickering
- Department of Molecular Medicine, UT Health San Antonio, Texas, TX 78245, USA;
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, Texas, TX 78245, USA
- The Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, UT Health San Antonio, TX 78229, USA
| | - Maria Gaczyńska
- Department of Molecular Medicine, UT Health San Antonio, Texas, TX 78245, USA;
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, Texas, TX 78245, USA
| |
Collapse
|
13
|
Lu BS, Yin YW, Zhang YP, Guo PY, Li W, Liu KL. Upregulation of NPL4 promotes bladder cancer cell proliferation by inhibiting DXO destabilization of cyclin D1 mRNA. Cancer Cell Int 2019; 19:149. [PMID: 31164795 PMCID: PMC6543671 DOI: 10.1186/s12935-019-0874-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 05/27/2019] [Indexed: 12/11/2022] Open
Abstract
Background NPL4 is an important cofactor of the valosin-containing protein (VCP)–NPL4–UFD1 complex. The VCP–NPL4–UFD1 has been considered as a ubiquitin proteasome system (UPS) regulator and response to protein degradation. While NPL4 plays important roles in various diseases, little is known about its functions in bladder cancer (BC). Methods MTT assays and colony forming test were performed to evaluate cell proliferation ability and Western blotting was used to detect protein expression. Cyclin D1 mRNA expression was detected using qRT-PCR, and coimmunoprecipitation (CoIP) was used to detect protein–protein interactions. Results NPL4 was upregulated in BC tissue and correlated with poor prognosis. Upregulation of NPL4 promoted cell proliferation while suppression of NPL4 reduced BC cell proliferation. Upregulation of NPL4 led to overexpression of cyclin D1 by enhancing its mRNA stability. Moreover, NPL4 was found to bind directly to DXO and induce its degradation. DXO was downregulated in BC tissue and regulated BC cell proliferation by destabilizing cyclin D1 mRNA. DXO-mediated NPL4 regulated BC cell proliferation by stabilizing cyclin D1 expression. Conclusions The NPL4/DXO/cyclin D1 axis exert crucial role in BC cell growth and is associated with prognosis and may represent a potential therapeutic target for BC.
Collapse
Affiliation(s)
- Bao-Sai Lu
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000 Hebei People's Republic of China
| | - Yue-Wei Yin
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000 Hebei People's Republic of China
| | - Yan-Ping Zhang
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000 Hebei People's Republic of China
| | - Ping-Ying Guo
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000 Hebei People's Republic of China
| | - Wei Li
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000 Hebei People's Republic of China
| | - Kai-Long Liu
- Department of Urology, The Second Hospital of Hebei Medical University, No. 215, Heping West Road, Shijiazhuang, 050000 Hebei People's Republic of China
| |
Collapse
|