Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.532
Filter
1.
Biomed Pharmacother ; 176: 116920, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38876054

ABSTRACT

Sarcopenia is a major public health concern among older adults, leading to disabilities, falls, fractures, and mortality. This study aimed to elucidate the pathophysiological mechanisms of sarcopenia and identify potential therapeutic targets using systems biology approaches. RNA-seq data from muscle biopsies of 24 sarcopenic and 29 healthy individuals from a previous cohort were analysed. Differential expression, gene set enrichment, gene co-expression network, and topology analyses were conducted to identify target genes implicated in sarcopenia pathogenesis, resulting in the selection of 6 hub genes (PDHX, AGL, SEMA6C, CASQ1, MYORG, and CCDC69). A drug repurposing approach was then employed to identify new pharmacological treatment options for sarcopenia (clofibric-acid, troglitazone, withaferin-a, palbociclib, MG-132, bortezomib). Finally, validation experiments in muscle cell line (C2C12) revealed MG-132 and troglitazone as promising candidates for sarcopenia treatment. Our approach, based on systems biology and drug repositioning, provides insight into the molecular mechanisms of sarcopenia and offers potential new treatment options using existing drugs.


Subject(s)
Drug Repositioning , Sarcopenia , Systems Biology , Humans , Sarcopenia/drug therapy , Sarcopenia/metabolism , Sarcopenia/genetics , Drug Repositioning/methods , Aged , Animals , Gene Regulatory Networks/drug effects , Male , Mice , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Female , Cell Line , Troglitazone , Molecular Targeted Therapy , Leupeptins/pharmacology , Leupeptins/therapeutic use
2.
Mol Biol Rep ; 51(1): 770, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38896168

ABSTRACT

BACKGROUND: MG132, a proteasome inhibitor, is widely used to inhibit nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activity by proteasome-mediated degradation of IκB. It has been marketed as a specific, reversible, cell-permeable and low-cost inhibitor. However, adverse effects of the compound have been reported in the literature. We recently discovered and characterised a point mutation in the acute phase protein serum amyloid A (SAA) in chickens, by overexpressing the protein in chicken hepatocellular carcinoma (LMH) cells. This serine to arginine exchange at amino acid position 90 (SAA.R90S) leads to intra- and extracellular accumulation of SAA, which is surprisingly counteracted by MG132 treatment, independent of SAA's intrinsic promoter. METHODS AND RESULTS: To test, whether low proteasomal degradation of SAA.R90S is responsible for the observed intra- and extracellular SAA accumulation, we intended to inhibit the proteasome in SAA wild type (SAA.WT) overexpressing cells with MG132. However, we observed an unexpected drastic decrease in SAA protein expression at the transcript level. NF-κB gene expression was unchanged by MG132 at the measured time point. CONCLUSIONS: The observed results demonstrate that MG132 inhibits SAA expression at the transcript level, independent of its endogenous promoter. Further, the data might indicate that NF-κB is not involved in the observed MG132-induced inhibition of SAA expression. We, consequently, question in this brief report whether MG132 should truly be categorised as a specific ubiquitin proteasome inhibitor and recommend the usage of alternative compounds.


Subject(s)
Carcinoma, Hepatocellular , Chickens , Leupeptins , Liver Neoplasms , NF-kappa B , Promoter Regions, Genetic , Serum Amyloid A Protein , Animals , Leupeptins/pharmacology , Chickens/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/drug therapy , Cell Line, Tumor , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/drug therapy , Promoter Regions, Genetic/genetics , Serum Amyloid A Protein/genetics , Serum Amyloid A Protein/metabolism , NF-kappa B/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteasome Endopeptidase Complex/genetics , Proteasome Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects
3.
Int J Mol Sci ; 25(10)2024 May 16.
Article in English | MEDLINE | ID: mdl-38791473

ABSTRACT

Reduced graphene oxide (rGO) and a proteasome inhibitor (MG-132) are some of the most commonly used compounds in various biomedical applications. However, the mechanisms of rGO- and MG-132-induced cytotoxicity remain unclear. The aim of this study was to investigate the anticancer effect of rGO and MG-132 against ZR-75-1 and MDA-MB-231 breast cancer cell lines. The results demonstrated that rGO, MG-132 or a mix (rGO + MG-132) induced time- and dose-dependent cytotoxicity in ZR-75-1 and MDA-MB-231 cells. Apart from that, we found that treatment with rGO and MG-132 or the mix increased apoptosis, necrosis and induction of caspase-8 and caspase-9 activity in both breast cancer cell lines. Apoptosis and caspase activation were accompanied by changes in the ultrastructure of mitochondria in ZR-75-1 and MDA-MB-231 cells incubated with rGO. Additionally, in the analyzed cells, we observed the induction of oxidative stress, accompanied by increased apoptosis and cell necrosis. In conclusion, oxidative stress induces apoptosis in the tested cells. At the same time, both mitochondrial and receptor apoptosis pathways are activated. These studies provided new information on the molecular mechanisms of apoptosis in the ZR-75-1 and MDA-MB-231 breast cancer cell lines.


Subject(s)
Apoptosis , Breast Neoplasms , Graphite , Oxidative Stress , Proteasome Inhibitors , Humans , Graphite/pharmacology , Graphite/chemistry , Apoptosis/drug effects , Oxidative Stress/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Proteasome Inhibitors/pharmacology , Female , Leupeptins/pharmacology , Drug Synergism , Mitochondria/drug effects , Mitochondria/metabolism
4.
COPD ; 21(1): 2342797, 2024 12.
Article in English | MEDLINE | ID: mdl-38712759

ABSTRACT

Objective: To investigate the effects of cigarette smoke (CS) on Serine/Threonine Kinase 11 (STK11) and to determine STK11's role in CS-induced airway epithelial cell cytotoxicity.Methods: STK11 expression levels in the lung tissues of smokers with or without COPD and mice exposed to CS or room air (RA) were determined by immunoblotting and RT-PCR. BEAS-2Bs-human bronchial airway epithelial cells were exposed to CS extract (CSE), and the changes in STK11 expression levels were determined by immunoblotting and RT-PCR. BEAS-2B cells were transfected with STK11-specific siRNA or STK11 expression plasmid, and the effects of CSE on airway epithelial cell cytotoxicity were measured. To determine the specific STK11 degradation-proteolytic pathway, BEAS-2Bs were treated with cycloheximide alone or combined with MG132 or leupeptin. Finally, to identify the F-box protein mediating the STK11 degradation, a screening assay was performed using transfection with a panel of FBXL E3 ligase subunits.Results: STK11 protein levels were significantly decreased in the lung tissues of smokers with COPD relative to smokers without COPD. STK11 protein levels were also significantly decreased in mouse lung tissues exposed to CS compared to RA. Exposure to CSE shortened the STK11 mRNA and protein half-life to 4 h in BEAS-2B cells. STK11 protein overexpression attenuated the CSE-induced cytotoxicity; in contrast, its knockdown augmented CSE-induced cytotoxicity. FBXL19 mediates CSE-induced STK11 protein degradation via the ubiquitin-proteasome pathway in cultured BEAS-2B cells. FBXL19 overexpression led to accelerated STK11 ubiquitination and degradation in a dose-dependent manner.Conclusions: Our results suggest that CSE enhances the degradation of STK11 protein in airway epithelial cells via the FBXL19-mediated ubiquitin-proteasomal pathway, leading to augmented cell death.HIGHLIGHTSLung tissues of COPD-smokers exhibited a decreased STK11 RNA and protein expression.STK11 overexpression attenuates CS-induced airway epithelial cell cytotoxicity.STK11 depletion augments CS-induced airway epithelial cell cytotoxicity.CS diminishes STK11 via FBXL19-mediated ubiquitin-proteasome degradation.


Subject(s)
AMP-Activated Protein Kinases , Epithelial Cells , F-Box Proteins , Protein Serine-Threonine Kinases , Smoke , Animals , Humans , Male , Mice , AMP-Activated Protein Kinase Kinases , Cell Line , Cigarette Smoking/adverse effects , Cycloheximide/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/drug effects , F-Box Proteins/metabolism , F-Box Proteins/genetics , Leupeptins/pharmacology , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Proteolysis/drug effects , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/genetics , Respiratory Mucosa/metabolism , Respiratory Mucosa/drug effects , RNA, Small Interfering , Smoke/adverse effects
5.
J Pharmacol Sci ; 155(2): 52-62, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677786

ABSTRACT

The ubiquitin-proteasome system (UPS) is a major proteolytic system that plays an important role in the regulation of various cell processes, such as cell cycle, stress response, and transcriptional regulation, especially in neurons, and dysfunction of UPS is considered to be a cause of neuronal cell death in neurodegenerative diseases. However, the mechanism of neuronal cell death caused by UPS dysfunction has not yet been fully elucidated. In this study, we investigated the mechanism of neuronal cell death induced by proteasome inhibitors using human neuroblastoma SH-SY5Y cells. Z-Leu-D-Leu-Leu-al (MG132), a proteasome inhibitor, induced apoptosis in SH-SY5Y cells in a concentration- and time-dependent manner. Antioxidants N-acetylcysteine and EUK-8 attenuated MG132-induced apoptosis. Apocynin and diphenyleneiodonium, inhibitors of NADPH oxidase (NOX), an enzyme that produces superoxide anions, also attenuated MG132-induced apoptosis. It was also found that MG132 treatment increased the expression of NOX5, a NOX family member, and that siRNA-mediated silencing of NOX5 and BAPTA-AM, which inhibits NOX5 by chelating calcium, suppressed MG132-induced apoptosis and production of reactive oxygen species in SH-SY5Y cells. These results suggest that MG132 induces apoptosis in SH-SY5Y cells through the production of superoxide anion by NOX5.


Subject(s)
Apoptosis , Leupeptins , NADPH Oxidase 5 , NADPH Oxidases , Neuroblastoma , Proteasome Inhibitors , Superoxides , Humans , Apoptosis/drug effects , Apoptosis/genetics , Proteasome Inhibitors/pharmacology , Superoxides/metabolism , Cell Line, Tumor , Neuroblastoma/pathology , Neuroblastoma/metabolism , Leupeptins/pharmacology , NADPH Oxidases/metabolism , NADPH Oxidases/genetics , NADPH Oxidase 5/genetics , NADPH Oxidase 5/metabolism , Antioxidants/pharmacology , Dose-Response Relationship, Drug , Acetylcysteine/pharmacology , Neurons/metabolism , Neurons/drug effects
6.
Cryobiology ; 115: 104882, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38452847

ABSTRACT

Oocyte cryopreservation is useful for human fertility treatment and strain preservation in both experimental and domestic animals. However, the embryonic development of vitrified rat oocytes was lower than that of vitrified embryos. To increase the viability of vitrified oocytes, intracellular ice formation during cooling and warming must be prevented. Rapid warming is important to prevent ice formation. Furthermore, suppressing the spontaneous activation of oocytes is also important because vitrification promotes the spontaneous activation of rat oocytes, and thus compromise developmental competence of the gametes. MG132, a proteasome inhibitor, suppresses the spontaneous activation of rat oocytes. Here, we examined the effects of rapid warming and MG132 treatment on the survival and embryonic development of vitrified rat oocytes. The warming rate was adjusted by changing the vitrification solution volume and warming solution temperature. The survival rate of oocytes vitrified in 10 µL solution and warmed at 50 °C (94%) was significantly higher than that of oocytes vitrified in 100 µL and 10 µL solution and warmed at 37 °C (49% and 81%, respectively). Furthermore, the rate of embryonic development of vitrified oocytes treated with MG132 during vitrification, warming, and intracytoplasmic sperm injection (ICSI) (44%) was significantly higher than that of untreated gametes (10%). Offspring were obtained after transferring embryos derived from MG132-treated vitrified oocytes (14%). Altogether, the survivability of vitrified rat oocytes increased by rapid warming, and MG132 improved embryonic development after ICSI.


Subject(s)
Cryopreservation , Embryonic Development , Leupeptins , Oocytes , Sperm Injections, Intracytoplasmic , Vitrification , Animals , Oocytes/drug effects , Oocytes/cytology , Rats , Female , Leupeptins/pharmacology , Cryopreservation/methods , Embryonic Development/drug effects , Sperm Injections, Intracytoplasmic/methods , Cell Survival/drug effects , Male , Cryoprotective Agents/pharmacology
7.
Cell Death Dis ; 13(10): 865, 2022 10 12.
Article in English | MEDLINE | ID: mdl-36224178

ABSTRACT

Acute-on-chronic liver failure is a distinct clinical syndrome characterized by a dysregulated immune response and extensive hepatocyte death without satisfactory therapies. As a cytoplasmic degradative and quality-control process, autophagy was implicated in maintaining intracellular homeostasis, and decreased hepatic autophagy was found in many liver diseases and contributes to disease pathogenesis. Previously, we identified the therapeutic potential of mesenchymal stem cells (MSCs) in ACLF patients; however, the intrinsic mechanisms are incompletely understood. Herein, we showed that MSCs restored the impaired autophagic flux and alleviated liver injuries in ACLF mice, but these effects were abolished when autophago-lysosomal maturation was inhibited by leupeptin (leu), suggesting that MSCs exerted their hepatoprotective function in a pro-autophagic dependent manner. Moreover, we described a connection between transcription factor EB (TFEB) and autophagic activity in this context, as evidenced by increased nuclei translocation of TFEB elicited by MSCs were capable of promoting liver autophagy. Mechanistically, we confirmed that let-7a-5p enriched in MSCs derived exosomes (MSC-Exo) could activate autophagy by targeting MAP4K3 to reduce TFEB phosphorylation, and MAP4K3 knockdown partially attenuates the effect of anti-let-7a-5p oligonucleotide via decreasing the inflammatory response, in addition, inducing autophagy. Altogether, these findings revealed that the hepatoprotective effect of MSCs may partially profit from its exosomal let-7a-5p mediating autophagy repairment, which may provide new insights for the therapeutic target of ACLF treatment.


Subject(s)
Acute-On-Chronic Liver Failure , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Mesenchymal Stem Cells , MicroRNAs/genetics , Acute-On-Chronic Liver Failure/genetics , Acute-On-Chronic Liver Failure/metabolism , Animals , Autophagy , Leupeptins/pharmacology , Mesenchymal Stem Cells/metabolism , Mice , Oligonucleotides/metabolism
8.
Physiol Rep ; 10(15): e15411, 2022 08.
Article in English | MEDLINE | ID: mdl-35924300

ABSTRACT

Prolonged tourniquet use can lead to tissue ischemia and can cause progressive muscle and nerve injuries. Such injuries are accompanied by calpain activation and subsequent Wallerian-like degeneration. Several known inhibitors, including leupeptin, are known to impede the activity of calpain and associated tissue damage. We hypothesize that employment of leupeptin in a rat model of prolonged hind limb ischemia can mitigate muscle and nerve injuries. Sprague-Dawley rats (n = 10) weighing between 300-400 g were employed in this study. Their left hind limbs were subjected to blood flow occlusion for a period of 2-h using a neonatal blood pressure cuff. Five rats were given twice weekly intramuscular leupeptin injections, while the other five received saline. After 2 weeks, the animals were euthanized, their sciatic nerves and gastrocnemius muscles were harvested, fixed, stained, and analyzed using NIH Image J software. The administration of leupeptin resulted in larger gastrocnemius muscle fiber cross-sectional areas for the right (non-tourniquet applied) hindlimb as compared to that treated with the saline (p = 0.0110). However, no statistically significant differences were found between these two groups for the injured left hindlimb (p = 0.1440). With regards to the sciatic nerve cross-sectional areas and sciatic functional index, no differences were detected between the leupeptin and control treated groups for both the healthy and injured hindlimbs. This research provides new insights on how to employ leupeptin to inhibit the degenerative effects of calpain and preserve tissues following ischemia resulting from orthopedic or plastic surgery procedures.


Subject(s)
Calpain , Ischemia , Animals , Hindlimb/blood supply , Ischemia/drug therapy , Leupeptins/pharmacology , Muscle, Skeletal , Rats , Rats, Sprague-Dawley
9.
Cells ; 11(4)2022 02 10.
Article in English | MEDLINE | ID: mdl-35203262

ABSTRACT

Progeroid syndromes (PS), including Hutchinson-Gilford Progeria Syndrome (HGPS), are premature and accelerated aging diseases, characterized by clinical features mimicking physiological aging. Most classical HGPS patients carry a de novo point mutation within exon 11 of the LMNA gene encoding A-type lamins. This mutation activates a cryptic splice site, leading to the production of a truncated prelamin A, called prelamin A ∆50 or progerin, that accumulates in HGPS cell nuclei and is a hallmark of the disease. Some patients with PS carry other LMNA mutations and are named "HGPS-like" patients. They produce progerin and/or other truncated prelamin A isoforms (∆35 and ∆90). We previously found that MG132, a proteasome inhibitor, induced progerin clearance in classical HGPS through autophagy activation and splicing regulation. Here, we show that MG132 induces aberrant prelamin A clearance and improves cellular phenotypes in HGPS-like patients' cells other than those previously described in classical HGPS. These results provide preclinical proof of principle for the use of a promising class of molecules toward a potential therapy for children with HGPS-like or classical HGPS.


Subject(s)
Progeria , Cell Nucleus , Humans , Leupeptins/pharmacology , Leupeptins/therapeutic use , Phenotype , Progeria/drug therapy , Progeria/genetics
10.
PLoS One ; 17(2): e0262612, 2022.
Article in English | MEDLINE | ID: mdl-35196318

ABSTRACT

Orthodontic treatment requires the regulation of bone remodeling in both compression and tension sides. Transforming growth factor-ß1 (TGF-ß1) is an important coupling factor for bone remodeling. However, the mechanism underlying the TGF-ß1-mediated regulation of the osteoclast-supporting activity of osteoblasts and stromal cells remain unclear. The current study investigated the effect of TGF-ß1 on receptor activator of nuclear factor kappa-B ligand (RANKL) expression in stromal cells induced by 1α,25(OH)2D3 (D3) and dexamethasone (Dex). TGF-ß1 downregulated the expression of RANKL induced by D3 and Dex in mouse bone marrow stromal lineage, ST2 cells. Co-culture system revealed that TGF-ß1 suppressed osteoclast differentiation from bone marrow cell induced by D3 and Dex-activated ST2 cells. The inhibitory effect of TGF-ß1 on RANKL expression was recovered by inhibiting the interaction between TGF-ß1 and the TGF-ß type I/activin receptor or by downregulating of smad2/3 expression. Interestingly, TGF-ß1 degraded the retinoid X receptor (RXR)-α protein which forms a complex with vitamin D receptor (VDR) and regulates transcriptional activity of RANKL without affecting nuclear translocation of VDR and phosphorylation of signal transducer and activator of transcription3 (STAT3). The degradation of RXR-α protein by TGF-ß1 was recovered by a ubiquitin-proteasome inhibitor. We also observed that poly-ubiquitination of RXR-α protein was induced by TGF-ß1 treatment. These results indicated that TGF-ß1 downregulates RANKL expression and the osteoclast-supporting activity of osteoblasts/stromal cells induced by D3 and Dex through the degradation of the RXR-α protein mediated by ubiquitin-proteasome system.


Subject(s)
Osteoclasts/drug effects , Osteoclasts/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction/drug effects , Transforming Growth Factor beta1/pharmacology , Ubiquitin/metabolism , Ubiquitination/drug effects , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Coculture Techniques , Leupeptins/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Osteoclasts/cytology , Proteasome Inhibitors/pharmacology , Recombinant Proteins/pharmacology , Signal Transduction/genetics , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Stromal Cells/drug effects , Stromal Cells/metabolism , Transfection , Ubiquitination/genetics
11.
Sci Rep ; 12(1): 2706, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35177721

ABSTRACT

Intracytoplasmic sperm injection (ICSI) is an effective reproductive technique for obtaining rat offspring using preserved sperm with low or no motility. However, rat oocytes undergo spontaneous activation immediately after retrieval from the oviduct and poorly develop after ICSI unless it is performed quickly. Here, we evaluated whether treatment with MG132, the proteasome inhibitor, suppresses the spontaneous activation of oocytes before and during ICSI. After retrieval from the oviducts, the rate of development into morula and blastocyst from the oocytes cultured in vitro for 1 h prior to ICSI significantly decreased compared with that from the control oocytes subject to ICSI without culture (7% versus 36%). However, a higher proportion of oocytes treated with MG132 for 0, 1, and 3 h before and during ICSI developed into morulae and blastocysts (70%, 60%, and 52%, respectively). Offspring were obtained from oocytes treated with MG132 for 0 and 1 h before and during ICSI (percentage: 31%). Altogether, MG132 could suppress the spontaneous activation of rat oocytes and increase embryonic development after ICSI.


Subject(s)
Embryonic Development/drug effects , Leupeptins/pharmacology , Leupeptins/therapeutic use , Oocytes/drug effects , Sperm Injections, Intracytoplasmic/methods , Animals , Chorionic Gonadotropin/pharmacology , Chorionic Gonadotropin/therapeutic use , Chromosomes/drug effects , Female , Male , Oocytes/cytology , Rats, Wistar , Sperm Injections, Intracytoplasmic/drug effects , Spermatozoa/drug effects , Time Factors
12.
PLoS Genet ; 18(1): e1010015, 2022 01.
Article in English | MEDLINE | ID: mdl-35025870

ABSTRACT

Oculopharyngeal muscular dystrophy (OPMD) is a late-onset disorder characterized by progressive weakness and degeneration of specific muscles. OPMD is due to extension of a polyalanine tract in poly(A) binding protein nuclear 1 (PABPN1). Aggregation of the mutant protein in muscle nuclei is a hallmark of the disease. Previous transcriptomic analyses revealed the consistent deregulation of the ubiquitin-proteasome system (UPS) in OPMD animal models and patients, suggesting a role of this deregulation in OPMD pathogenesis. Subsequent studies proposed that UPS contribution to OPMD involved PABPN1 aggregation. Here, we use a Drosophila model of OPMD to address the functional importance of UPS deregulation in OPMD. Through genome-wide and targeted genetic screens we identify a large number of UPS components that are involved in OPMD. Half dosage of UPS genes reduces OPMD muscle defects suggesting a pathological increase of UPS activity in the disease. Quantification of proteasome activity confirms stronger activity in OPMD muscles, associated with degradation of myofibrillar proteins. Importantly, improvement of muscle structure and function in the presence of UPS mutants does not correlate with the levels of PABPN1 aggregation, but is linked to decreased degradation of muscle proteins. Oral treatment with the proteasome inhibitor MG132 is beneficial to the OPMD Drosophila model, improving muscle function although PABPN1 aggregation is enhanced. This functional study reveals the importance of increased UPS activity that underlies muscle atrophy in OPMD. It also provides a proof-of-concept that inhibitors of proteasome activity might be an attractive pharmacological approach for OPMD.


Subject(s)
Muscular Atrophy/pathology , Muscular Dystrophy, Oculopharyngeal/pathology , Poly(A)-Binding Protein I/genetics , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/metabolism , Animals , Disease Models, Animal , Drosophila melanogaster , Gene Expression Regulation , Genetic Testing , Humans , Leupeptins/pharmacology , Leupeptins/therapeutic use , Muscular Atrophy/drug therapy , Muscular Atrophy/metabolism , Muscular Dystrophy, Oculopharyngeal/drug therapy , Muscular Dystrophy, Oculopharyngeal/genetics , Muscular Dystrophy, Oculopharyngeal/metabolism , Mutation , Poly(A)-Binding Protein I/chemistry , Proof of Concept Study , Protein Aggregates/drug effects
13.
J Bacteriol ; 204(1): e0045621, 2022 01 18.
Article in English | MEDLINE | ID: mdl-34633870

ABSTRACT

The protective mechanisms of blood-brain barrier (BBB) prohibiting entry of pathogens into central nervous system (CNS) are critical for maintenance of brain homeostasis. These include various intracellular defense mechanisms that are vital to block transcytosis of neurotropic pathogens into the CNS. However, mechanistic details of coordination between these defense pathways remain unexplored. In this study, we established that BBB-driven ubiquitination acts as a major intracellular defense mechanism for clearance of Streptococcus pneumoniae, a critical neurotropic pathogen, during transit through BBB. Our findings suggest that the BBB employs differential ubiquitination with either K48- or K63-ubiquitin (Ub) chain topologies as an effective strategy to target S. pneumoniae toward diverse killing pathways. While K63-Ub decoration triggers autophagic killing, K48-Ub directs S. pneumoniae exclusively toward proteasomes. Time-lapse fluorescence imaging involving proteasomal marker LMP2 revealed that in the BBB, the majority of the ubiquitinated S. pneumoniae was cleared by proteasome. Fittingly, inhibition of proteasome and autophagy pathway led to accumulation of K48-Ub- and K63-Ub-marked S. pneumoniae, respectively, and triggered significant increases in intracellular S. pneumoniae burden. Moreover, genetic impairment of either K48- or K63-Ub chain formation demonstrated that although both chain types are key in disposal of intracellular S. pneumoniae, K48-Ub chains and subsequent proteasomal degradation have more pronounced contributions to intracellular S. pneumoniae killing in the BBB. Collectively, these observations, for the first time, illustrated a pivotal role of differential ubiquitination deployed by BBB in orchestrating a symphony of intracellular defense mechanisms for interception and degradation of S. pneumoniae, blocking its entry into the brain, which could be exploited to prevent bacterial CNS infections. IMPORTANCE The blood-brain barrier (BBB) represents a unique cellular barrier that provides structural integrity and protection to the CNS from pathogen invasion. Recently, ubiquitination, which is key for cellular homeostasis, was shown to be involved in pathogen clearance. In this study, we deciphered that the BBB deploys differential ubiquitination as an effective strategy to prevent S. pneumoniae trafficking into the brain. The different ubiquitin chain topologies formed on S. pneumoniae dictated the selection of downstream degradative pathways, namely, autophagy and proteasomes, among which the contribution of the proteasomal system in S. pneumoniae killing is more pronounced. Overall our study revealed how the BBB deploys differential ubiquitination as a strategy for synchronization of various intracellular defense pathways, which work in tandem to ensure the brain's identity as an immunologically privileged site.


Subject(s)
Blood-Brain Barrier/physiology , Endothelial Cells/physiology , Gene Expression Regulation, Bacterial/physiology , Streptococcus pneumoniae/physiology , Ubiquitins/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacology , Autophagy/drug effects , Biomarkers , Cell Line , Cell Survival/drug effects , Gene Expression Regulation, Bacterial/drug effects , Gentamicins/administration & dosage , Gentamicins/pharmacology , Humans , Leupeptins/pharmacology , Optical Imaging/methods , Penicillins/administration & dosage , Penicillins/pharmacology , Proteasome Endopeptidase Complex/metabolism , Ubiquitination , Ubiquitins/chemistry
14.
Microvasc Res ; 140: 104276, 2022 03.
Article in English | MEDLINE | ID: mdl-34742813

ABSTRACT

PURPOSE: We previously reported that a calpain inhibitor (CAI) prevents the development of atherosclerosis in rats. This study aimed to investigate the effects of CAI (1 mg/kg) on atherosclerosis in apolipoprotein E knockout (ApoE KO) mice that were fed a high-fat diet (HFD) and explore the underlying mechanism by analyzing the expression of genes related to the uptake and efflux of cholesterol. METHODS: Atherosclerotic plaques were evaluated. The activity of calpain in the aorta and that of superoxide dismutase (SOD) in the serum were assessed. Lipid profiles in the serum and liver were examined. Serum oxidized low-density lipoprotein (oxLDL), malondialdehyde (MDA), tumor necrosis factor (TNF-α), and interleukin-6 (IL-6) levels were measured. The mRNA expressions of CD68, TNF-α, IL-6, CD36, scavenger receptor (SR-A), peroxisome proliferator-activated receptor gamma (PPAR-γ), liver-x-receptor alpha (LXR-α), and ATP-binding cassette transporter class A1 (ABCA1) in the aorta and peritoneal macrophages were also evaluated. RESULTS: CAI reduced calpain activity in the aorta. CAI also impeded atherosclerotic lesion formation and mRNA expression of CD68 in the aorta and peritoneal macrophages of ApoE KO mice compared with those of mice receiving HFD. However, CAI had no effect on body weight and lipid levels in both the serum and liver. CAI significantly decreased MDA, oxLDL, TNF-α, and IL-6 levels and increased SOD activity in the serum. Moreover, CAI significantly inhibited the mRNA expression of TNF-α and IL-6 genes in the aorta and peritoneal macrophages. In addition, CAI significantly downregulated the mRNA expression of scavenger receptors CD36 and SR-A and upregulated the expression of genes involved in the cholesterol efflux pathway, i.e., PPAR-γ, LXR-α, and ABCA1 in the aorta and peritoneal macrophages. CONCLUSIONS: CAI inhibited the development of atherosclerotic lesions in ApoE KO mice, and this effect might be related to the reduction of oxidative stress and inflammation and the improvement of cholesterol intake and efflux pathways.


Subject(s)
Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Calpain/antagonists & inhibitors , Cholesterol/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Leupeptins/pharmacology , Lipid Metabolism/drug effects , Macrophages, Peritoneal/drug effects , RNA, Messenger/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/genetics , Antigens, Differentiation, Myelomonocytic/metabolism , Aorta/enzymology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Calpain/metabolism , Disease Models, Animal , Gene Expression Regulation , Lipid Metabolism/genetics , Liver X Receptors/genetics , Liver X Receptors/metabolism , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/pathology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , PPAR gamma/genetics , PPAR gamma/metabolism , Plaque, Atherosclerotic , RNA, Messenger/genetics , Scavenger Receptors, Class A/genetics , Scavenger Receptors, Class A/metabolism
15.
J Appl Microbiol ; 132(2): 1176-1184, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34496097

ABSTRACT

AIMS: Effects of a proteasome inhibitor, MG-132, on the riboflavin production in Ashbya gossypii were investigated to elucidate the relationship of the riboflavin production with flavoprotein homeostasis. METHODS AND RESULTS: The addition of MG-132 to the liquid medium reduced the specific riboflavin production by 79% in A. gossypii at 25 µM after 24 h. The addition of the inhibitor also caused the accumulation of reactive oxygen species and ubiquitinated proteins. These results indicated that MG-132 works in A. gossypii without any genetic engineering and reduces riboflavin production. In the presence of 25 µM MG-132, specific NADH dehydrogenase activity was increased by 1.4-fold compared to DMSO, but specific succinate dehydrogenase (SDH) activity was decreased to 52% compared to DMSO. Additionally, the amount of AgSdh1p (ACR052Wp) was also reduced. Specific riboflavin production was reduced to 22% when 20 mM malonate, a SDH inhibitor, was added to the culture medium. The riboflavin production in heterozygous AgSDH1 gene-disrupted mutant (AgSDH1-/+ ) was reduced to 63% compared to that in wild type. CONCLUSIONS: MG-132 suppresses the riboflavin production and SDH activity in A. gossypii. SDH is one of the flavoproteins involved in the riboflavin production in A. gossypii. SIGNIFICANCE AND IMPACT OF THE STUDY: This study shows that MG-132 has a negative influence on the riboflavin production and SDH activity in A. gossypii and leads to the elucidation of the connection of the riboflavin production with flavoproteins.


Subject(s)
Proteasome Inhibitors , Riboflavin , Saccharomycetales/metabolism , Genetic Engineering , Leupeptins/pharmacology , Proteasome Inhibitors/pharmacology , Riboflavin/biosynthesis , Saccharomycetales/drug effects
16.
Bioengineered ; 13(2): 3620-3633, 2022 02.
Article in English | MEDLINE | ID: mdl-34699308

ABSTRACT

Preeclampsia (PE) is a pregnancy disorder characterized by excessive trophoblast cell death. This study aims to explore the exact mechanism of the ubiquitination level of FUN14 domain containing 1 (FUNDC1) in mitophagy and injury in hypoxic trophoblast cells. In this study, HTR-8/SVneo trophoblast cells were cultured under normoxic and hypoxic conditions and PE mouse model was established. We found low ubiquitination level of FUNDC1 in hypoxic trophoblast cells and placenta of pregnant women with PE. Proteasome inhibitor MG-132 and protease activator MF-094 were added into HTR-8/SVneo trophoblast cells. Proteasome inhibitor MG-132 decreased FUNDC1 ubiquitination level while protease activator MF-094 increased FUNDC1 ubiquitination level. Inhibition of FUNDC1 ubiquitination promoted mitophagy and mitochondrial membrane potential (Δψm) in normoxic trophoblast cells, increased levels of reactive oxygen species (ROS) and malondialdehyde (MDA) and decreased levels of glutathione (GSH) and superoxide dismutase (SOD). In addition, FUNDC1 ubiquitination alleviated cell injury in PE mice in vivo. In conclusion, increased FUNDC1 ubiquitination level inhibited mitophagy and Δψm changes in hypoxic trophoblast cells, and thus alleviated oxidative injury.


Subject(s)
Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Mitophagy , Pre-Eclampsia/metabolism , Trophoblasts/metabolism , Ubiquitination , Cell Hypoxia/drug effects , Cell Line , Female , Humans , Leupeptins/pharmacology , Pregnancy
17.
Int J Mol Sci ; 22(21)2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34769210

ABSTRACT

After almost two years from its first evidence, the COVID-19 pandemic continues to afflict people worldwide, highlighting the need for multiple antiviral strategies. SARS-CoV-2 main protease (Mpro/3CLpro) is a recognized promising target for the development of effective drugs. Because single target inhibition might not be sufficient to block SARS-CoV-2 infection and replication, multi enzymatic-based therapies may provide a better strategy. Here we present a structural and biochemical characterization of the binding mode of MG-132 to both the main protease of SARS-CoV-2, and to the human Cathepsin-L, suggesting thus an interesting scaffold for the development of double-inhibitors. X-ray diffraction data show that MG-132 well fits into the Mpro active site, forming a covalent bond with Cys145 independently from reducing agents and crystallization conditions. Docking of MG-132 into Cathepsin-L well-matches with a covalent binding to the catalytic cysteine. Accordingly, MG-132 inhibits Cathepsin-L with nanomolar potency and reversibly inhibits Mpro with micromolar potency, but with a prolonged residency time. We compared the apo and MG-132-inhibited structures of Mpro solved in different space groups and we identified a new apo structure that features several similarities with the inhibited ones, offering interesting perspectives for future drug design and in silico efforts.


Subject(s)
COVID-19 Drug Treatment , Cathepsin L/drug effects , Coronavirus 3C Proteases/drug effects , Leupeptins/chemistry , Leupeptins/pharmacology , SARS-CoV-2/chemistry , SARS-CoV-2/drug effects , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , Catalytic Domain/drug effects , Cathepsin L/chemistry , Coronavirus 3C Proteases/chemistry , Drug Design , Drug Discovery , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Peptidomimetics , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Virus Replication/drug effects , X-Ray Diffraction
18.
Cell Death Dis ; 12(10): 937, 2021 10 13.
Article in English | MEDLINE | ID: mdl-34645792

ABSTRACT

Breast cancer is the most commonly diagnosed malignant tumor among females. Estrogen receptor α (ERα) is initially expressed in 70% of breast cancers and is a well-known target of endocrine therapy for ERα-positive breast cancer. In the present study, we identified MINDY1, a member belongs to the motif interacting with Ubcontaining novel DUB family (MINDY), as a potential deubiquitylase of ERα in breast cancer. There was a positive correlation between ERα and MINDY1 protein levels in human breast cancer tissues. We found that high expression of MINDY1 was associated with poor prognosis. MINDY1 interacted with ERα, thereby mediating the deubiquitination of ERα and increased its stability in a deubiquitylation activity-dependent manner. MINDY1 depletion significantly decreased the ERα protein level and ERα signaling activity in breast cancer cells. Specifically, MINDY1 associated with the N-terminal of ERα via its catalytic domain, thus inhibiting K48-specific poly-ubiquitination process on ERα protein. In addition, MINDY1 depletion led to growth inhibition and cell cycle arrest of ERα-positive breast cancer cells. Finally, overexpression of ERα could rescue the MINDY1 depletion-induced growth inhibition both in vitro and in vivo, suggesting that MINDY1 promotes breast carcinogenesis through increasing ERα stability. Overall, our study proposed a novel post-translational mechanism of ERα in supporting breast cancer progression. Targeting the MINDY1 may prove to be a promising strategy for patients with ERα-positive breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Deubiquitinating Enzymes/metabolism , Estrogen Receptor alpha/metabolism , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , HEK293 Cells , Humans , Leupeptins/pharmacology , Mice, Inbred BALB C , Mice, Nude , Prognosis , Proteasome Inhibitors/pharmacology , Protein Binding/drug effects , Protein Stability/drug effects , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Ubiquitination/drug effects
19.
Int J Mol Sci ; 22(19)2021 Sep 30.
Article in English | MEDLINE | ID: mdl-34638951

ABSTRACT

The protein phosphorylation of the membrane-bound mitochondrial proteins has become of interest from the point of view of its regulatory role of the function of the respiratory chain, opening of the mitochondrial permeability transition pore (mPTP), and initiation of apoptosis. Earlier, we noticed that upon phosphorylation of proteins in some proteins, the degree of their phosphorylation increases with the opening of mPTP. Two isoforms of myelin basic protein and cyclic nucleotide phosphodiesterase were identified in rat brain non-synaptic mitochondria and it was concluded that they are involved in mPTP regulation. In the present study, using the mass spectrometry method, the phosphorylated protein was identified as Calpain 3 in rat brain non-synaptic mitochondria. In the present study, the phosphoprotein Calpain-3 (p94) (CAPN3) was identified in the rat brain mitochondria as a phosphorylated truncated form of p60-62 kDa by two-dimensional electrophoresis and mass spectrometry. We showed that the calpain inhibitor, calpeptin, was able to suppress the Ca2+ efflux from mitochondria, preventing the opening of mPTP. It was found that phosphorylated truncated CALP3 with a molecular weight of 60-62 contains p-Tyr, which indicates the possible involvement of protein tyrosine phosphatase in this process.


Subject(s)
Brain/metabolism , Calcium Signaling/drug effects , Calpain/metabolism , Isoenzymes/metabolism , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Permeability Transition Pore/metabolism , Muscle Proteins/metabolism , Animals , Apoptosis/drug effects , Calcium/metabolism , Calpain/antagonists & inhibitors , Calpain/chemistry , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Leupeptins/pharmacology , Male , Molecular Weight , Muscle Proteins/antagonists & inhibitors , Muscle Proteins/chemistry , Phosphorylation , Protein Transport , Rats
20.
J Neuroimmunol ; 361: 577752, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34715591

ABSTRACT

Neuromyelitis Optica (NMO) is an autoimmune inflammatory disease that affects the optic nerves and spinal cord. The autoantibody is generated against the abundant water channel protein of the brain, Aquaporin 4 (AQP4). Of the two isoforms of AQP4, the shorter one (M23) often exists as a supramolecular assembly known as an orthogonal array of particles (OAPs). There have been debates about the fate of these AQP4 clusters upon binding to the antibody, the exact mechanism of its turnover, and the proteins associated with the process. Recently several clinical cases of NMO were reported delineating the effect of Rituximab (RTX) therapy. Extending these reports at the cell signaling level, we developed a glioma based cellular model that mimicked antibody binding and helped us track the subsequent events including a variation of AQP4 levels, alterations in cellular morphology, and the changes in downstream signaling cascades. Our results revealed the extent of perturbations in the signaling pathways related to stress involving ERK, JNK, and AKT1 together with markers for cell death. We could also decipher the possible routes of degradation of AQP4, post-exposure to antibody. We further investigated the effect of autoantibody on AQP4 transcriptional level and involvement of FOXO3a and miRNA-145 in the regulation of transcription. This study highlights the differential outcome at the cellular level when treated with the serum of the same patient pre and post RTX therapy and for the first time mechanistically describes the effect of RTX.


Subject(s)
Aquaporin 4/metabolism , Autoantibodies/blood , Autoantigens/metabolism , Immunoglobulin G/blood , Neuromyelitis Optica/metabolism , Rituximab/therapeutic use , Adult , Aquaporin 4/genetics , Aquaporin 4/immunology , Autoantigens/genetics , Autoantigens/immunology , Cell Line, Tumor , Cell Membrane/chemistry , Cell Membrane/ultrastructure , Cell Shape , Cell Surface Extensions/ultrastructure , Female , Forkhead Box Protein O3/physiology , Glioblastoma , Humans , Leupeptins/pharmacology , Male , MicroRNAs/genetics , Microscopy, Confocal , Neuromyelitis Optica/drug therapy , Neuromyelitis Optica/immunology , Proteasome Endopeptidase Complex/metabolism , Signal Transduction , Single-Cell Analysis , Transcription, Genetic , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...