80 results on '"Proteasome pathway"'
Search Results
2. Genome-wide identification of PYL/PYR-PP2C (A)-SnRK2 genes in Eutrema and their co-expression analysis in response to ABA and abiotic stresses
- Author
-
Li, Chuanshun, Zhang, Hengyang, Qi, Yuting, Zhao, Yaoyao, Duan, Chonghao, Wang, Yujiao, Meng, Zhe, and Zhang, Quan
- Published
- 2023
- Full Text
- View/download PDF
3. Heat shock protein (HSP90) affects the replication of rotavirus.
- Author
-
Hu, Siman, Tao, Xiao-Li, Zhao, Wei, Luo, Like, Li, Runli, Ma, Zhuping, Sun, Lvyin, and Li, Yong-Gang
- Abstract
Aim: To explore the effects of HSP90 on rotavirus (RV) replication. Materials & methods: The interaction between NSP2 and HSP90 was analyzed by immunoprecipitation assay, and co-localization confirmed by an immunofluorescence assay. The viral titer was measured by immunofluorescence assay and viral protein expression investigated by western blotting. Results: The interaction between NSP2 and HSP90 was confirmed. Knockdown of HSP90 using siRNA resulted in a decrease in NSP2 expression and virus titer. HSP90 inhibitor 17-AAG significantly decreased NSP2 and NSP5 expression, as well as the virus titer, while proteasome inhibitor MG132 restored the protein amount and the viral titer. Conclusion: HSP90 is involved in rotavirus replication, and inhibition of HSP90 promotes the degradation of NSP2 through the proteasome pathway. Rotavirus is a type of virus that causes severe diarrhea, especially in children. The virus infects the cell in our bodies. These cells have proteins which carry out important functions and can help the growth of the virus. One of these proteins, called HSP90, interacts with a protein of the virus, called NSP2. After infection, these proteins move to the same place within the cell. Decreasing the level of HSP90 also decreases the level of NSP2, slowing the growth of the virus. Understanding how HSP90 and NSP2 work will help to develop new drugs to treat rotavirus infection. Article highlights Background Heat shock protein 90 (HSP90) appears to play an essential role in viral replication, but little is known regarding the role of HSP90 in rotavirus (RV) infection. Methods RV laboratory strain SA11 was used in this study. Virus titers were measured by Fluorescent formation units assay and viral protein expression checked by WB. The interaction of HSP90 and NSP2 was confirmed by immunoprecipitation (IP) and confocal laser microscope. siRNA used to knockdown the expression of HSP90 and HSP90 activity was inhibited. Results The HSP90 inhibition resulted in decreased RV replication. Knockdown of HSP90 by siRNA decreased RV protein levels and virus replication. HSP90 interacted with virus protein NSP2. Inhibition of the proteasome pathway decreased viral protein degradation. Conclusion HSP90 was used by RV to facilitate virus replication by chaperoning viral proteins and preventing their degradation via the proteasome. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Proteomic signature associated with chronic kidney disease (CKD) progression identified by data-independent acquisition mass spectrometry.
- Author
-
Ramírez Medina, Carlos R., Ali, Ibrahim, Baricevic-Jones, Ivona, Odudu, Aghogho, Saleem, Moin A., Whetton, Anthony D., Kalra, Philip A., and Geifman, Nophar
- Subjects
- *
PROTEOLYSIS , *CHRONIC kidney failure , *MASS spectrometry , *PROTEOMICS , *DIABETIC nephropathies , *COMPLEMENT inhibition - Abstract
Background: Halting progression of chronic kidney disease (CKD) to established end stage kidney disease is a major goal of global health research. The mechanism of CKD progression involves pro-inflammatory, pro-fibrotic, and vascular pathways, but pathophysiological differentiation is currently lacking. Methods: Plasma samples of 414 non-dialysis CKD patients, 170 fast progressors (with ∂ eGFR-3 ml/min/1.73 m2/year or worse) and 244 stable patients (∂ eGFR of − 0.5 to + 1 ml/min/1.73 m2/year) with a broad range of kidney disease aetiologies, were obtained and interrogated for proteomic signals with SWATH-MS. We applied a machine learning approach to feature selection of proteins quantifiable in at least 20% of the samples, using the Boruta algorithm. Biological pathways enriched by these proteins were identified using ClueGo pathway analyses. Results: The resulting digitised proteomic maps inclusive of 626 proteins were investigated in tandem with available clinical data to identify biomarkers of progression. The machine learning model using Boruta Feature Selection identified 25 biomarkers as being important to progression type classification (Area Under the Curve = 0.81, Accuracy = 0.72). Our functional enrichment analysis revealed associations with the complement cascade pathway, which is relevant to CKD as the kidney is particularly vulnerable to complement overactivation. This provides further evidence to target complement inhibition as a potential approach to modulating the progression of diabetic nephropathy. Proteins involved in the ubiquitin–proteasome pathway, a crucial protein degradation system, were also found to be significantly enriched. Conclusions: The in-depth proteomic characterisation of this large-scale CKD cohort is a step toward generating mechanism-based hypotheses that might lend themselves to future drug targeting. Candidate biomarkers will be validated in samples from selected patients in other large non-dialysis CKD cohorts using a targeted mass spectrometric analysis. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
5. Dabie bandavirus Nonstructural Protein Interacts with Actin to Induce F-Actin Rearrangement and Inhibit Viral Adsorption and Entry.
- Author
-
Hongyun Liu, Sihua Liu, Zixiang Liu, Xiaoning Gao, Leling Xu, Mengqian Huang, Yazhi Su, Zhiyun Wang, and Tao Wang
- Subjects
- *
CYTOSKELETON , *F-actin , *ACTIN , *CELL aggregation , *CLATHRIN , *ENDOCYTOSIS , *CELLULAR inclusions , *UBIQUITINATION - Abstract
Dabie bandavirus (DBV) is an emerging Bandavirus that causes multiorgan failure with a high fatality rate in humans. While many viruses can manipulate the actin cytoskeleton to facilitate viral growth, the regulation pattern of the actin cytoskeleton and the molecular mechanisms involved in DBV entry into the host cells remain unclear. In this study, we demonstrate that expression of nonstructural protein (NSs) or infection with DBV induces actin rearrangement, which presents a point-like distribution, and this destruction is dependent on inclusion bodies (IBs). Further experiments showed that NSs inhibits viral adsorption by destroying the filopodium structure. In addition, NSs also compromised the viral entry by inhibiting clathrin aggregation on the cell surface and capturing clathrin into IBs. Furthermore, NSs induced clathrin light chain B (CLTB) degradation through the K48-linked ubiquitin proteasome pathway, which could negatively regulate clathrin-mediated endocytosis, inhibiting the viral entry. Finally, we confirmed that this NSs-induced antiviral mechanism is broadly applicable to other viruses, such as enterovirus 71 (EV71) and influenza virus, A/PR8/34 (PR8), which use the same clathrin-mediated endocytosis to enter host cells. In conclusion, our study provides new insights into the role of NSs in inhibiting endocytosis and a novel strategy for treating DBV infections. IMPORTANCE Dabie bandavirus (DBV), a member of the Phenuiviridae family, is a newly emerging tick-borne pathogen that causes multifunctional organ failure and even death in humans. The actin cytoskeleton is involved in various crucial cellular processes and plays an important role in viral life activities. However, the relationship between DBV infection and the actin cytoskeleton has not been described in detail. Here, we show for the first time the interaction between NSs and actin to induce actin rearrangement, which inhibits the viral adsorption and entry. We also identify a key mechanism underlying NSs-induced entry inhibition in which NSs prevents clathrin aggregation on the cell surface by hijacking clathrin into the inclusion body and induces CLTB degradation through the K48-linked ubiquitination modification. This paper is the first to reveal the antiviral mechanism of NSs and provides a theoretical basis for the search for new antiviral targets. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
6. ATP citrate lyase promotes the progression of hepatocellular carcinoma by activating the REGγ-proteasome pathway.
- Author
-
Cai Q, Zhu H, Dai Y, Zhou Q, Zhang Q, and Zhu Q
- Subjects
- Animals, Humans, Mice, Cell Line, Tumor, Autoantigens metabolism, Autoantigens genetics, Signal Transduction, Antigens, Neoplasm metabolism, Antigens, Neoplasm genetics, Male, Carcinoma, Hepatocellular pathology, Carcinoma, Hepatocellular genetics, Carcinoma, Hepatocellular metabolism, Liver Neoplasms pathology, Liver Neoplasms genetics, Liver Neoplasms metabolism, ATP Citrate (pro-S)-Lyase metabolism, ATP Citrate (pro-S)-Lyase genetics, Proteasome Endopeptidase Complex metabolism, Proteasome Endopeptidase Complex genetics, Cell Proliferation, Gene Expression Regulation, Neoplastic, Cell Movement, Disease Progression
- Abstract
The search for novel tumor biomarkers and targets is of significant importance for the early clinical diagnosis and treatment of Hepatocellular Carcinoma (HCC). The mechanisms by which ATP citrate lyase (ACLY) promotes HCC progression remain unclear, and the connection between ACLY and REGγ has not been reported in the literature. In vitro, we will perform overexpression/knockdown of ACLY or overexpression/knockdown of REGγ to investigate the impact of ACLY on HCC cells and its underlying mechanisms. In vivo, we will establish mouse tumor models with overexpression/knockdown of ACLY or overexpression/knockdown of REGγ to study the effect of ACLY on mouse tumors and its mechanisms. Firstly, ACLY overexpression upregulated REGγ expression and activated the REGγ-proteasome pathway, leading to changes in the expression of downstream signaling pathway proteins. This promoted HCC cell proliferation, invasion, and migration in vitro, as well as tumor growth and metastasis in vivo. Secondly, ACLY overexpression increased acetyl-CoA production, upregulated the acetylation level of the REGγ promoter region histone H3K27ac, and subsequently induced REGγ expression. Lastly, enhanced acetylation of the REGγ promoter region histone H3K27ac resulted in upregulated REGγ expression, activation of the REGγ-proteasome pathway, changes in downstream signaling pathway protein expression, and promotion of HCC cell proliferation, invasion, and migration in vitro, as well as tumor growth and metastasis in vivo. Conversely, REGγ knockdown reversed these effects. ACLY and REGγ may serve as potential biomarkers and clinical therapeutic targets for HCC., (© 2024 Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
7. Mechanisms underlying extensive Ser129-phosphorylation in α-synuclein aggregates
- Author
-
Shigeki Arawaka, Hiroyasu Sato, Asuka Sasaki, Shingo Koyama, and Takeo Kato
- Subjects
Parkinson’s disease ,α–Synuclein ,Phosphorylation ,Aggregation ,Mitochondrial impairment ,Proteasome pathway ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Parkinson’s disease (PD) is characterized neuropathologically by intracellular aggregates of fibrillar α-synuclein, termed Lewy bodies (LBs). Approximately 90% of α-synuclein deposited as LBs is phosphorylated at Ser129 in brains with PD. In contrast, only 4% of total α-synuclein is phosphorylated at Ser129 in brains with normal individuals. It is unclear why extensive phosphorylation occurs in the pathological process of PD. To address this issue, we investigated a mechanism and role of Ser129-phosphorylation in regulating accumulation of α-synuclein. In CHO cells, the levels of Ser129-phosphorylated soluble α-synuclein were maintained constantly to those of total α-synuclein in intracellular and extracellular spaces. In SH-SY5Y cells and rat primary cortical neurons, mitochondrial impairment by rotenone or MPP+ enhanced Ser129-phosphorylation through increased influx of extracellular Ca2+. This elevation was suppressively controlled by targeting Ser129-phosphorylated α-synuclein to the proteasome pathway. Rotenone-induced insoluble α-synuclein was also targeted by Ser129-phosphoryation to the proteasome pathway. Experiments with epoxomicin and chloroquine showed that proteasomal targeting of insoluble Ser129-phosphorylated α-synuclein was enhanced under lysosome inhibition and it reduced accumulation of insoluble total α-synuclein. However, in a rat AAV-mediated α-synuclein overexpression model, there was no difference in the number of total α-synuclein aggregates between A53T mutant and A53T plus S129A double mutant α-synuclein, although Ser129-phosphorylated α-synuclein-positive aggregates were increased in rats expressing A53T α-synuclein. These findings suggest that Ser129-phosphorylation occurs against stress conditions, which increases influx of extracellular Ca2+, and it prevents accumulation of insoluble α-synuclein by evoking proteasomal clearance complementary to lysosomal one. However, Ser129-phosphorylation may provide an ineffective signal for degradation-resistant aggregates, causing extensive phosphorylation in aggregates.
- Published
- 2017
- Full Text
- View/download PDF
8. Sclerostin inhibits odontogenic differentiation of human pulp‐derived odontoblast‐like cells under mechanical stress.
- Author
-
Liao, Chufang, Ou, Yanjing, Wu, Yun, Zhou, Yi, Liang, Shanshan, and Wang, Yining
- Subjects
- *
SCLEROSTIN , *RUNX proteins , *OSTEOCALCIN , *PROTEASOME inhibitors - Abstract
Sclerotic dentin is a natural self‐protective barrier beneath non‐carious cervical lesions (NCCLs), which are mainly induced by mechanical stress. Sclerostin is a mechanosensory protein and serves as an inhibitor of dentinogenesis. However, its function on mechanotransduction in dentine–pulp complex has not been elucidated yet. In this study, decreased sclerostin expression was detected in odontoblasts beneath NCCL‐affected sclerotic dentin. Then human pulp‐derived odontoblast‐like cells (hOBs) were subjected to mechanical strain (MS) in vitro: the results showed that MS‐induced upregulation of odontogenic differentiation markers (dentin sialophosphoprotein, osteopontin, osteocalcin, and runt‐related transcription factor 2) in hOBs with downregulated sclerostin expression, and this inductive differentiation was attenuated when sclerostin was overexpressed. Additionally, MS activated ERK1/2 pathway and ERK1/2 inhibition restored MS‐induced downregulation of sclerostin. Proteasome inhibitor MG132 could also rescue MS‐induced decrease of sclerostin. Furthermore, MS suppressed STAT3 pathway, which could be reversed by sclerostin overexpression. STAT3 inhibition was shown to ameliorate the reduction of odontogenic markers induced by sclerostin overexpression. Taken together, we conclude that MS downregulates sclerostin expression via the ERK1/2 and proteasome signaling pathways to promote odontogenic differentiation of hOBs through the STAT3 signaling pathway. It can therefore be inferred that under mechanical stress, sclerostin inhibition promotes reactive dentin formation by enhancing odontogenic differentiation of odontoblasts, which might be one of potential forming mechanisms of sclerotic dentin beneath NCCLs. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
9. Bortezomib’s Scientific Origins and Its Tortuous Path to the Clinic
- Author
-
Goldberg, Alfred L., Ghobrial, Irene M., editor, Richardson, Paul G., editor, and Anderson, Kenneth C., editor
- Published
- 2011
- Full Text
- View/download PDF
10. Mechanisms underlying extensive Ser129-phosphorylation in a-synuclein aggregates.
- Author
-
Shigeki Arawaka, Hiroyasu Sato, Asuka Sasaki, Shingo Koyama, and Takeo Kato
- Subjects
PARKINSON'S disease ,SYNUCLEINS ,PHOSPHORYLATION ,MITOCHONDRIAL pathology ,PROTEASOMES - Abstract
Parkinson's disease (PD) is characterized neuropathologically by intracellular aggregates of fibrillar α-synuclein, termed Lewy bodies (LBs). Approximately 90% of α-synuclein deposited as LBs is phosphorylated at Ser129 in brains with PD. In contrast, only 4% of total α-synuclein is phosphorylated at Ser129 in brains with normal individuals. It is unclear why extensive phosphorylation occurs in the pathological process of PD. To address this issue, we investigated a mechanism and role of Ser129-phosphorylation in regulating accumulation of α-synuclein. In CHO cells, the levels of Ser129-phosphorylated soluble α-synuclein were maintained constantly to those of total α- synuclein in intracellular and extracellular spaces. In SH-SY5Y cells and rat primary cortical neurons, mitochondrial impairment by rotenone or MPP
+ enhanced Ser129-phosphorylation through increased influx of extracellular Ca2+ . This elevation was suppressively controlled by targeting Ser129-phosphorylated α-synuclein to the proteasome pathway. Rotenone-induced insoluble α-synuclein was also targeted by Ser129-phosphoryation to the proteasome pathway. Experiments with epoxomicin and chloroquine showed that proteasomal targeting of insoluble Ser129-phosphorylated α-synuclein was enhanced under lysosome inhibition and it reduced accumulation of insoluble total α-synuclein. However, in a rat AAV-mediated α-synuclein overexpression model, there was no difference in the number of total α-synuclein aggregates between A53T mutant and A53T plus S129A double mutant α-synuclein, although Ser129-phosphorylated α-synuclein-positive aggregates were increased in rats expressing A53T α-synuclein. These findings suggest that Ser129-phosphorylation occurs against stress conditions, which increases influx of extracellular Ca2+ , and it prevents accumulation of insoluble α-synuclein by evoking proteasomal clearance complementary to lysosomal one. However, Ser129-phosphorylation may provide an ineffective signal for degradation-resistant aggregates, causing extensive phosphorylation in aggregates. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
11. Trametinib potentiates TRAIL‐induced apoptosis via FBW7‐dependent Mcl‐1 degradation in colorectal cancer cells
- Author
-
Penglong Cao, Bing Li, Dapeng Ding, Shijun Li, Xiaoguang Xiao, and Lin Lin
- Subjects
0301 basic medicine ,Proteasome Endopeptidase Complex ,F-Box-WD Repeat-Containing Protein 7 ,Pyridones ,Colorectal cancer ,Down-Regulation ,TRAIL ,Pyrimidinones ,TNF-Related Apoptosis-Inducing Ligand ,03 medical and health sciences ,0302 clinical medicine ,Ubiquitin ,Trametinib ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Humans ,Phosphorylation ,neoplasms ,degradation ,Proteasome Pathway ,Glycogen Synthase Kinase 3 beta ,biology ,Chemistry ,apoptosis ,Ubiquitination ,Original Articles ,Cell Biology ,medicine.disease ,Ubiquitin ligase ,030104 developmental biology ,Mcl‐1 ,Apoptosis ,030220 oncology & carcinogenesis ,Proteolysis ,biology.protein ,Cancer research ,Myeloid Cell Leukemia Sequence 1 Protein ,Molecular Medicine ,Original Article ,Colorectal Neoplasms ,Protein Binding - Abstract
Trametinib is a MEK1/2 inhibitor and exerts anticancer activity against a variety of cancers. However, the effect of Trametinib on colorectal cancer (CRC) is not well understood. In the current study, our results demonstrate the ability of sub‐toxic doses of Trametinib to enhance TRAIL‐mediated apoptosis in CRC cells. Our findings also indicate that Trametinib and TRAIL activate caspase‐dependent apoptosis in CRC cells. Moreover, Mcl‐1 overexpression can reduce apoptosis in CRC cells treated with Trametinib with or without TRAIL. We further demonstrate that Trametinib degrades Mcl‐1 through the proteasome pathway. In addition, GSK‐3β phosphorylates Mcl‐1 at S159 and promotes Mcl‐1 degradation. The E3 ligase FBW7, known to polyubiquitinate Mcl‐1, is involved in Trametinib‐induced Mcl‐1 degradation. Taken together, these results provide the first evidence that Trametinib enhances TRAIL‐mediated apoptosis through FBW7‐dependent Mcl‐1 ubiquitination and degradation.
- Published
- 2020
- Full Text
- View/download PDF
12. Investigation of resistance mechanisms developed against proteasome inhibitors in human prostate cancer cell line PC3
- Author
-
Kanbur, Ertan, Budak, Ferah, Yerlikaya, Azmi, and Bursa Uludağ Üniversitesi/Sağlık Bilimleri Enstitüsü/Tıp Fakültesi/İmmünoloji Anabilim Dalı
- Subjects
Bortezomib ,İlaç direnci mekanizmaları ,Drug resistance mechanisms ,Proteasome pathway ,Proteozom yolağı ,Proteozom inhibitörleri ,Senesens ,Proteasome inhibitors ,Kanser ,Senescence ,Cancer - Abstract
Günümüzde kanser tedavisi için en umut vaat eden uygulama halen kemoterapi olmasına rağmen kanser ilaçlarına karşı ilaç direnci, medikal onkolojideki en büyük engel teşkil etmeye devam etmektedir. Kemoterapide meydana gelen başarısızlık sebeplerinin %90’ı, kanserde olan veya oluşan ilaç direnci ile meydana gelen invazyon ve metastazdan kaynaklanmaktadır. Tüm dünyada prostat kanseri, erkeklerde akciğer kanserinden (%14,3) sonra ikinci (%14,1) en sık görülen kanser tipidir. Ubiquitin ve proteozom yolağı (UPY) proteinler için bir kalite kontrol sistemidir. Hasarlı ve yanlış katlanmış proteinlerin yıkımından sorumludur. UPY; transkripsiyonda, hücre döngüsünde, hücre içi sinyallerde, antijen sunumunda ve epigenetik mekanizmalarda görev almaktadır. Proteozom inhibitörleri hematolojik malignitelerde kullanılmaktadır. Bortezomib bir proteozom inhibitörüdür. Proteozom inhibitörlerinin solid tümörlerde kullanılması umut vadetmektedir. Proteozom inhibitörlerinin otoimmün hastalıklarda da kullanılabileceği düşünülmektedir. Hücrelerin stres kaynaklarına verdiği yanıtlardan biri, hücrelerdeki moleküler değişiklikler ile kalıcı bir şekilde hücre döngüsü tutuklanması olan senesenstir. Senesent hücreler genellikle biyoaktif proteinler salgılarlar ve bunlar senesent olmayan komşu hücreleri uyararak tümör gelişimine sebep de olabilmektedir ve aynı zamanda yaşlanma sürecindeki kronik enflamasyondan sorumlu olduğu düşünülmektedir. Bu tezde; bir prostat kanseri hücre hattı olan PC3 hücrelerinde [PC3-P, parental; PC3-R, dirençli], klinikte prostat tedavi protokollerinde henüz kullanılmayan ve solid tümörlerde kullanılması umut vadeden bortezomib ilacına karşı oluşmuş ilaç direnci mekanizmaları ile bortezomib’in PC3 hücreleri üzerindeki senesens rolü araştırılmıştır. Dirençli PC3-R ve parental PC3-P hücrelerinde yaptığımız bortezomib sitotoksisitesi, hücre ölüm modu, AO/EB çift boyaması ve 3 boyutlu hücre kültürü çalışmalarında; PC3-R hücrelerinin PC3-P hücrelerine göre ilaca çok daha dirençli oldukları, apoptoza daha dirençli oldukları ve ilaç varlığında bile 3 boyutlu kompleks yapıları oluşturarak kendilerini destekleyebildikleri ve direnç mekanizmasının geri dönüşümlü olmadığı gösterilmiştir. Direnç mekanizmasını aydınlatmaya yönelik MAPK ve otofajik aktivasyon, ısı şok proteinlerin ekspresyonu gibi incelenen stratejik yolaklarda; stres koşullarında aktive olan ve dirence sebebiyet veren bu yolakların ve ilgili moleküllerin dirençli PC3-R hücrelerinde değil, parental PC3-P hücrelerinde bir sağkalım mekanizması olarak aktive oldukları gözlenmiştir. PC3-R hücrelerindeki bortezomib direncinin ilacın bağlandığı PSMB5 enziminde meydana gelen bir mutasyondan kaynaklanabileceği düşünülmektedir. PC3-P ve PC3-R hücrelerinde yapılan senesens deneyleri sonucunda, senesens markerlerinin bortezomib muamelesi sonucunda ekseriyetle azaldığı gözlenmiştir ve bu sebeple bortezomibin bir senolitik etkisinin olabileceği öne sürülmektedir. Buna ek olarak PC3-R hücrelerinde senesens, PC3-P hücrelerine göre daha az görülmektedir ve bunun ilaç direncine bir katkısı olabilir. Son olarak, literatüre göre ERK1 ve ERK2 proteinlerinin fonksiyonel olarak işlevlerinin aynı olup olmadığı hakkında tartışmalar vardır. Tez çalışmamızda bortezomib muamelesinden sonra ERK1 fosforilasyonlarının artarken ERK2 fosforilasyonlarının azaldığı görülmüştür. Bu olay, ERK1 ve ERK2’nin farklı regüle edilebildiğini ve bu sebeple farklı işlevler görebileceklerine işaret etmektedir. Chemotherapy is still the most promising application for cancer treatment today. However, drug resistance to cancer drugs remains the biggest obstacle in medical oncology as 90% of the causes of failure in chemotherapy are due to invasion and metastasis caused by drug resistance in cancer. Prostate cancer is the second most common type of cancer in men (14.1%) following lung cancer (14.3%) in the world. Ubiquitin and the proteasome pathway (UPP) is a quality control system for proteins. It is responsible for the destruction of damaged and misfolded proteins. UPP is involved in transcription, cell cycle, intracellular signals, antigen presentation, and epigenetic mechanisms. Proteasome inhibitors are used in hematological malignancies. Bortezomib is a proteasome inhibitor. The use of proteasome inhibitors in solid tumors is promising. It is thought that proteasome inhibitors can also be used in autoimmune diseases. One of the cells' responses to stressors is senescence, which is a permanent cell cycle arrest by molecular changes in the cells. Senescent cells often secrete bioactive proteins, which can stimulate neighboring non-senescent cells to cause tumorigenesis and are also thought to be responsible for chronic inflammation in the aging process. Bortezomib is not yet used in prostate treatment protocols in clinics and bortezomib is promising to be used in solid tumors. In this thesis; drug resistance mechanisms against bortezomib resistance and the senescence role of bortezomib on PC3 cells were investigated in a prostate cancer cell line PC3 cells [PC3-P, parental; PC3-R, resistant]. In the studies of bortezomib cytotoxicity, cell death mode, AO/EB double staining, and 3D cell culture performed in resistant PC3-R and parental PC3-P cells; PC3-R cells were found to be much more resistant to bortezomib and apoptosis than PC3-P cells. We have shown that the PC3-R cells can support themselves by forming 3-dimensional complex structures even in the presence of bortezomib and the resistance mechanism in resistant PC3-R cells is not reversible. In the strategic pathways we examined, such as MAPK and autophagic activation, the expression of heat shock proteins for elucidating the resistance mechanism; it has been demonstrated that these pathways and their related molecules, which are activated under stress conditions and cause resistance, are more strongly activated in parental PC3-P cells and not in resistant PC3-R cells due to cell survival mechanisms. We think that bortezomib resistance may be due to a mutation in the PSMB5 enzyme to which bortezomib drug binds in the PC3-R cells. As a result of the senescence experiments performed in PC3-P and PC3-R cells, senescence markers were observed to be decreased to bortezomib treatment in general, and we, therefore, suggest that bortezomib may have a senolytic effect. Moreover, it has been observed that senescence is less common in PC3-R cells than in PC3-P cells, and this may contribute to drug resistance. Finally, according to the literature, there are debates about whether the ERK1 and ERK2 proteins are functionally the same. In this thesis study, it was observed that ERK1 phosphorylations increased while ERK2 phosphorylations decreased after bortezomib treatment. This indicates that ERK1 and ERK2 can be regulated differently and hence they may possess distinct functions.
- Published
- 2022
13. Bortezomib-mediated downregulation of S-phase kinase protein-2 (SKP2) causes apoptotic cell death in chronic myelogenous leukemia cells.
- Author
-
Iskandarani, Ahmad, Bhat, Ajaz A., Siveen, Kodappully S., Prabhu, Kirti S., Kuttikrishnan, Shilpa, Khan, Muzammil A., Krishnankutty, Roopesh, Kulinski, Michal, Nasr, Rihab R., Mohammad, Ramzi M., and Uddin, Shahab
- Subjects
- *
BORTEZOMIB , *MYELOID leukemia , *CELL death , *DRUG administration , *MULTIPLE myeloma , *PATIENTS , *PROTEIN metabolism , *ANTINEOPLASTIC agents , *APOPTOSIS , *BIOCHEMISTRY , *CARRIER proteins , *CELL cycle , *CELL lines , *CELL physiology , *PHENOMENOLOGY , *MITOCHONDRIA , *PROTEOLYTIC enzymes , *PROTEASE inhibitors , *CHRONIC myeloid leukemia , *PHARMACODYNAMICS - Abstract
Background: Proteasome inhibitors are attractive cancer therapeutic agents because they can regulate apoptosis-related proteins. Bortezomib also known as Velcade(®), a proteasome inhibitor that has been approved by the food and drug administration for treatment of patients with multiple myeloma, and many clinical trials are ongoing to examine to the efficacy of bortezomib for the treatment of other malignancies. Bortezomib has been shown to induce apoptosis and inhibit cell growth of many cancer cells. In current study, we determine whether bortezomib induces cell death/apoptosis in CML.Methods: Cell viability was measured using MTT assays. Apoptosis was measured by annexin V/PI dual staining and DNA fragmentation assays. Immunoblotting was performed to examine the expression of proteins. Colony assays were performed using methylcellulose.Results: Treatment of CML cells with bortezomib results in downregulation of S-phase kinase protein 2 (SKP2) and concomitant stabilization of the expression of p27Kip1. Furthermore, knockdown of SKP2 with small interference RNA specific for SKP2 caused accumulation of p27Kip1. CML cells exposed to bortezomib leads to conformational changes in Bax protein, resulting in loss of mitochondrial membrane potential and leakage of cytochrome c to the cytosol. In the cytosol, cytochrome c causes sequential activation of caspase-9, caspase-3, PARP cleavage and apoptosis. Pretreatment of CML cells with a universal inhibitor of caspases, z-VAD-fmk, prevents bortezomib-mediated apoptosis. Our data also demonstrated that bortezomib treatment of CML downregulates the expression of inhibitor of apoptosis proteins. Finally, inhibition of proteasome pathways by bortezomib suppresses colony formation ability of CML cells.Conclusions: Altogether, these findings suggest that bortezomib suppresses the cell proliferation via induction of apoptosis in CML cells by downregulation of SKP2 with concomitant accumulation of p27Kip1, suggesting that proteasomal pathway may form novel therapeutic targets for better management of CML. [ABSTRACT FROM AUTHOR]- Published
- 2016
- Full Text
- View/download PDF
14. Repair or Degrade: the Thermodynamic Dilemma of Cellular Protein Quality-Control
- Author
-
Bruno Fauvet, Mathieu E. Rebeaud, Satyam Tiwari, Paolo De Los Rios, and Pierre Goloubinoff
- Subjects
QH301-705.5 ,substrate ,Review ,Protein degradation ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Biochemistry ,Ribosome ,thermodynamics ,ATP hydrolysis ,chaperone system ,ubiquitin ,Native state ,chaperones ,Molecular Biosciences ,Biology (General) ,Molecular Biology ,proteostasis ,Chemistry ,heat-shock proteins ,complete genome sequence ,protein repair ,peptide ,Proteostasis ,proteasome pathway ,protein degradation ,Protein repair ,escherichia-coli ,Biophysics ,amino-acid ,Protein folding ,dnak ,Macromolecule - Abstract
Life is a non-equilibrium phenomenon. Owing to their high free energy content, the macromolecules of life tend to spontaneously react with ambient oxygen and water and turn into more stable inorganic molecules. A similar thermodynamic picture applies to the complex shapes of proteins: While a polypeptide is emerging unfolded from the ribosome, it may spontaneously acquire secondary structures and collapse into its functional native conformation. The spontaneity of this process is evidence that the free energy of the unstructured state is higher than that of the structured native state. Yet, under stress or because of mutations, complex polypeptides may fail to reach their native conformation and form instead thermodynamically stable aggregates devoid of biological activity. Cells have evolved molecular chaperones to actively counteract the misfolding of stress-labile proteins dictated by equilibrium thermodynamics. HSP60, HSP70 and HSP100 can inject energy from ATP hydrolysis into the forceful unfolding of stable misfolded structures in proteins and convert them into unstable intermediates that can collapse into the native state, even under conditions inauspicious for that state. Aggregates and misfolded proteins may also be forcefully unfolded and degraded by chaperone-gated endo-cellular proteases, and in eukaryotes also by chaperone-mediated autophagy, paving the way for their replacement by new, unaltered functional proteins. The greater energy cost of degrading and replacing a polypeptide, with respect to the cost of its chaperone-mediated repair represents a thermodynamic dilemma: some easily repairable proteins are better to be processed by chaperones, while it can be wasteful to uselessly try recover overly compromised molecules, which should instead be degraded and replaced. Evolution has solved this conundrum by creating a host of unfolding chaperones and degradation machines and by tuning their cellular amounts and activity rates.
- Published
- 2021
- Full Text
- View/download PDF
15. Endoplasmic reticulum stress and proteasome pathway involvement in human podocyte injury with a truncated COL4A3 mutation
- Author
-
Hui-Di Zhang, Jian-Ni Huang, Yun-Zi Liu, Hong Ren, Jing-Yuan Xie, Nan Chen, and Yuan-Yuan Ji
- Subjects
Collagen Type IV ,Proteasome Endopeptidase Complex ,MG132 ,Leupeptins ,Mutant ,Mutation, Missense ,lcsh:Medicine ,Biology ,urologic and male genital diseases ,medicine.disease_cause ,Autoantigens ,Frameshift mutation ,Podocyte ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Proteasome pathway ,medicine ,Animals ,Humans ,Missense mutation ,COL4A3 mutation ,Frameshift Mutation ,Mice, Knockout ,Collagen type IV-related nephropathy ,Mutation ,Brefeldin A ,Podocytes ,Endoplasmic reticulum ,Lentivirus ,lcsh:R ,Podocyte injury ,Original Articles ,General Medicine ,Endoplasmic Reticulum Stress ,Molecular biology ,medicine.anatomical_structure ,chemistry ,030220 oncology & carcinogenesis ,Proteasome inhibitor ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Background: Collagen type IV (COL4)-related nephropathy includes a variety of kidney diseases that occur with or without extra-renal manifestations caused by COL4A3-5 mutations. Previous studies revealed several novel mutations, including three COL4A3 missense mutations (G619R, G801R, and C1616Y) and the COL4A3 chr:228172489delA c.4317delA p.Thr1440ProfsX87 frameshift mutation that resulted in a truncated NC1 domain (hereafter named COL4A3 c.4317delA); however, the mutation mechanisms that lead to podocyte injury remain unclear. This study aimed to further explore the mutation mechanisms that lead to podocyte injury. Methods: Wild-type (WT) and four mutant COL4A3 segments were constructed into a lentiviral plasmid, then stably transfected into human podocytes. Real-time polymerase chain reaction and Western blotting were applied to detect endoplasmic reticulum stress (ERS)- and apoptosis-related mRNA and protein levels. Then, human podocytes were treated with MG132 (a proteasome inhibitor) and brefeldin A (a transport protein inhibitor). The human podocyte findings were verified by the establishment of a mus-Col4a3 knockout mouse monoclonal podocyte using clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9) technology. Results: Our data showed that COL4A3 mRNA was significantly overexpressed in the lentivirus stably transfected podocytes. Moreover, the COL4A3 protein level was significantly increased in all groups except the COL4A3 c.4317delA group. Compared to the other test groups, the COL4A3 c.4317delA group showed excessive ERS and apoptosis. Podocytes treated with MG132 showed remarkably increased intra-cellular expression of the COL4A3 c.4317delA mutation. MG132 intervention improved higher ERS and apoptosis levels in the COL4A3 c.4317delA group. Mouse monoclonal podocytes with COL4A3 chr:82717932insA c.4852insA p.Arg1618ThrfsX4 were successfully acquired; this NC1-truncated mutation suggested a higher level of ERS and relatively remarkable level of apoptosis compared to that of the WT group. Conclusions: We demonstrated that excessive ERS and ERS-induced apoptosis were involved in the podocyte injury caused by the NC1-truncated COL4A3 mutation. Furthermore, proteasome pathway intervention might become a potential treatment for collagen type IV-related nephropathy caused by a severely truncated COL4A3 mutation. Key words: Collagen type IV-related nephropathy; COL4A3 mutation; Podocyte injury; Proteasome pathway; MG132; Endoplasmic reticulum stress
- Published
- 2019
- Full Text
- View/download PDF
16. Genome wide analysis implicates upregulation of proteasome pathway in major depressive disorder
- Author
-
Uri Nitzan, Noam Shomron, Ifat Israel-Elgali, David Gurwitz, Yuval Bloch, Shaked Belaish, Israel Krieger, Michael Majer, Abraham Weizman, Libi Hertzberg, Guy Shapira, and Aviv Segev
- Subjects
Genetics ,Proteasome Pathway ,Depressive Disorder, Major ,Proteasome Endopeptidase Complex ,Depression ,Genome wide analysis ,Neurosciences. Biological psychiatry. Neuropsychiatry ,Biology ,medicine.disease ,Polymorphism, Single Nucleotide ,Article ,Up-Regulation ,Cellular and Molecular Neuroscience ,Psychiatry and Mental health ,Downregulation and upregulation ,medicine ,Major depressive disorder ,Humans ,Genetic Predisposition to Disease ,Biological Psychiatry ,Depression (differential diagnoses) ,RC321-571 ,Neuroscience ,Genome-Wide Association Study - Published
- 2021
17. Multiple lysines combined in HIV-1 Vif determines the responsiveness to CBF-β.
- Author
-
Ai, Youwei and Ma, Jianzhang
- Subjects
- *
PHYSIOLOGICAL effects of lysine , *HIV protease inhibitors , *VIRAL proteins , *PROTEASOME inhibitors , *PROMOTERS (Genetics) - Abstract
The Vif (viral infectivity factor) protein of human immunodeficiency virus type-1 (HIV-1) is critical for HIV-1 infectivity. CBF-β is required for HIV-1 Vif function, as it increases the steady-state level of the HIV-1 Vif protein to promote host restriction factor APOBEC3 degradation. However, the precise mechanism by which CBF-β promotes HIV-1 Vif levels remains unclear. In the present study, we provided evidences that CBF-β promoted steady-state levels of HIV-1 Vif by inhibiting the degradation of HIV-1 Vif through the proteasome pathway. Our results reveal a new mechanism by which a cellular protein supports viral infectivity by inhibiting viral protein degradation. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
18. Protein-protein interaction analysis of distinct molecular pathways in two subtypes of colorectal carcinoma.
- Author
-
HANZHANG CHEN, YUNZHEN FANG, HAILONG ZHU, SHUAI LI, TAO WANG, PAN GU, XIA FANG, YUNJIN WU, JUN LIANG, YU ZENG, LONG ZHANG, WEIZHE QIU, LANJING ZHANG, and XIANGHUA YI
- Subjects
- *
CARCINOMA , *PROTEIN-protein interactions , *GENE expression , *TUMOR necrosis factors , *NETWORK analysis (Planning) - Abstract
The aim of this study was to identify the molecular events that distinguish serrated colorectal carcinoma (SCRC) from conventional colorectal carcinoma (CCRC) through differential gene expression, pathway and protein-protein interaction (PPI) network analysis. The GSE4045 and GSE8671 microarray datasets were downloaded from the Gene Expression Omnibus database. We identified the genes that are differentially expressed between SCRC and normal colon tissues, CCRC and healthy tissues, and between SCRC and CCRC using Student's t-tests and Benjamini-Hochberg (BH) multiple testing corrections. The differentially expressed genes (DEGs) were then mapped to Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and their enrichment for specific pathways was investigated using the Database for Annotation, Visualization and Integrated Discovery (DAVID) tool with a significance threshold of 0.1. Analysis of the potential interactions between the protein products of 220 DEGs (between CCRC and SCRC) was performed by constructing a PPI network using data from the high performance RDF database (P<0.1). The interaction between pathways was also analyzed in CCRC based on the PPI network. Our study identified thousands of genes differentially expressed in SCRC and CCRC compared to healthy tissues. The DEGs in SCRC and CCRC were enriched in cell cycle, DNA replication, and base excision repair pathways. The proteasome pathway was significantly enriched in SCRC but not in CCRC after BH adjustment. The PPI network showed that tumour necrosis factor receptor-associated factor 6 (TRAF6) and atrophin 1 (ATN1) were the most central genes in the network, with respective degrees of node predicted at 90 and 88. In conclusion, the preoteasome pathway was shown to be specifically enriched in SCRC. Furthermore, TRAF6 and ATN1 may be promising biomarkers for the distinction between serrated and conventional CRC. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
19. Oroxylin A alleviates immunoparalysis of CLP mice by degrading CHOP through interacting with FBXO15
- Author
-
Ri Zhou, Yating Shan, Wu Yin, Zhaoxin Zhang, and Yun Wang
- Subjects
Male ,0301 basic medicine ,Inflammatory response ,Immunology ,lcsh:Medicine ,Diseases ,Pharmacology ,CHOP ,Article ,Sepsis ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Downregulation and upregulation ,Animals ,Medicine ,lcsh:Science ,Flavonoids ,Proteasome Pathway ,Multidisciplinary ,biology ,Drug discovery ,business.industry ,F-Box Proteins ,lcsh:R ,Cecal ligation ,medicine.disease ,Ubiquitin ligase ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Proteolysis ,biology.protein ,Oroxylin A ,lcsh:Q ,business ,Transcription Factor CHOP - Abstract
Clinical reports have found that with the improvement of treatment, most septic patients are able to survive the severe systemic inflammatory response and to enter the immunoparalysis stage. Considering that immunoparalysis leads to numerous deaths of clinical sepsis patients, alleviation of the occurrence and development of immunoparalysis has become a top priority in the treatment of sepsis. In our study, we investigate the effects of oroxylin A on sepsis in cecal ligation and puncture (CLP) mice. We find that the 60 h + 84 h (30 mg/kg) injection scheme of oroxylin A induce the production of pro-inflammatory factors, and further significantly improves the survival of CLP mice during the middle or late stages of sepsis. Mechanistically, C/EBP-homologous protein (CHOP) is upregulated and plays anti-inflammatory roles to facilitate the development of immunoparalysis in CLP mice. Oroxylin A induces the transcription of E3 ligase F-box only protein 15 gene (fbxo15), and activated FBXO15 protein binds to CHOP and further mediates the degradation of CHOP through the proteasome pathway, which eventually relieves the immunoparalysis of CLP mice. Taken together, these findings suggest oroxylin A relieves the immunoparalysis of CLP mice by degrading CHOP through interacting with FBXO15.
- Published
- 2020
- Full Text
- View/download PDF
20. Shifting the paradigm in treating multi-factorial diseases: polypharmacological co-inhibitors of HDAC6
- Author
-
Steven Fletcher and Alexandria M Chan
- Subjects
0301 basic medicine ,Pharmacology ,Drug ,Proteasome Pathway ,Combination therapy ,business.industry ,media_common.quotation_subject ,Organic Chemistry ,Pharmaceutical Science ,Disease ,HDAC6 ,Bioinformatics ,Biochemistry ,03 medical and health sciences ,Chemistry ,030104 developmental biology ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Drug Discovery ,Molecular Medicine ,Medicine ,Polypharmacology ,business ,Patient compliance ,media_common - Abstract
Multi-factorial diseases are illnesses that exploit multiple cellular processes, or stages within one process, and thus highly targeted therapies often succumb to the disease, losing efficacy as resistance sets in. Combination therapies have become a mainstay to battle these diseases, however these regimens are plagued with caveats. An emerging avenue to treat multi-factorial diseases is polypharmacology, wherein a single drug is rationally designed to bind multiple targets, and is widely touted to be superior to combination therapy by inherently addressing the latter's shortcomings, which include poor patient compliance, narrow therapeutic windows and spiraling healthcare costs. Through its roles in intracellular trafficking, cell motility, mitosis, protein folding and as a back-up to the proteasome pathway, HDAC6 has rapidly become an exciting new target for therapeutics, particularly in the discovery of new drugs to treat Alzheimer's disease and cancer. Herein, we describe recent efforts to marry together HDAC pharmacophores, with a particular emphasis on HDAC6 selectivity, with those of other targets towards the discovery of potent therapeutics to treat these evasive diseases. Such polypharmacological agents may supercede combination therapies through inherent synergism, permitting reduced dosing, wider therapeutic windows and improved compliance.
- Published
- 2020
21. SARS coronavirus 8b reduces viral replication by down-regulating E via an ubiquitin-independent proteasome pathway
- Author
-
Keng, Choong-Tat, Åkerström, Sara, Leung, Cynthia Sau-Wai, Poon, Leo L.M., Peiris, J.S. Malik, Mirazimi, Ali, and Tan, Yee-Joo
- Subjects
- *
SARS disease , *CORONAVIRUS diseases , *VIRAL replication , *UBIQUITIN , *GENE expression , *GENETIC regulation , *LYSINE , *PROTEINS - Abstract
Abstract: The severe acute respiratory syndrome coronavirus (SARS-CoV) 8b protein, which is not expressed by other known coronaviruses, can down-regulate the envelope (E) protein via a proteasome-dependent pathway. Here, we showed that the down-regulation of E is not dependent on the lysine residues on 8b and the reduction of polyubiquitination of E mutants is not correlated with their down-regulation by 8b, suggesting an ubiquitin-independent proteasome pathway is involved. A time-course study revealed that 8b was expressed at late-stages of SARS-CoV infection. By using Vero E6 cells stably expressing green fluorescence protein-tagged 8b, ectopic expression of 8b was shown to significantly reduce the production of progeny virus and down-regulate E expression. Taken together, these results suggest that 8b negatively modulates virus replication by down-regulating E via an ubiquitin-independent proteasome pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
22. Regulation of NF-κB in Human Vascular Tissue
- Author
-
Marczin, Nandor, Hoare, Ginette, Morrison, Karen, Chester, Adrian, Yacoub, Magdi, Catravas, John D., editor, Callow, Allan D., editor, and Ryan, Una S., editor
- Published
- 1998
- Full Text
- View/download PDF
23. A viral activator of gene expression functions via the ubiquitin-proteasome pathway.
- Author
-
Everett, Roger D., Orr, Anne, and Preston, Chris M.
- Subjects
- *
SKIN infections , *GENOMES , *GENETICS , *HERPES simplex virus , *UBIQUITIN , *GENE expression - Abstract
The ability of herpes simplex virus type 1 (HSV-1) to attain a latent state in sensory neurones and reactivate periodically is crucial for its biological and clinical properties. The active transcription of the entire 152 kb viral genome during lytic replication contrasts with the latent state, which is characterized by the production of a single set of nuclear-retained transcripts. Reactivation of latent genomes to re-initiate the lytic cycle therefore involves a profound change in viral transcriptional activity, but the mechanisms by which this fundamentally important process occurs are yet to be well understood. In this report we show that the stimulation of the onset of viral lytic infection mediated by the viral immediate-early (IE) protein Vmw110 is strikingly inhibited by inactivation of the ubiquitin—proteasome pathway. Similarly, the Vmw110-dependent reactivation of quiescent viral genomes in cultured cells is also dependent on proteasome activity. These results constitute the first demonstration that the transcriptional activity of a viral genome can be regulated by protein stability control pathways. [ABSTRACT FROM AUTHOR]
- Published
- 1998
- Full Text
- View/download PDF
24. Inhibition of the DNA-Sensing pathway by pseudorabies virus UL24 protein via degradation of interferon regulatory factor 7.
- Author
-
Liu, Xuelan, Zhang, Mingliang, Ye, Chao, Ruan, Keyue, Xu, Aiyun, Gao, Fei, Tong, Guangzhi, and Zheng, Hao
- Subjects
- *
INTERFERON regulatory factors , *TYPE I interferons , *AUJESZKY'S disease virus , *VIRAL proteins , *PROTEOLYSIS , *DNA virus diseases , *INTERFERONS - Abstract
• Pseudorabies Virus UL24 inhibited IFN-β. • UL24 antagonized cGAS-STING signal pathway. • UL24 interacted with and degraded IRF7 via the proteasome pathway. The cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) signaling pathway plays an important role in the innate immune response by the production of type I interferon (IFN) against DNA virus infection. However, viruses have evolved a variety of strategies to antagonize the host antiviral response to facilitate infection and replication. Pseudorabies virus (PRV), a DNA virus that causes great economic losses to the swine industry, encodes approximate 70 proteins, including some that are involved in evasion of host immunity. However, the mechanism employed by PRV to regulate type I IFN remains unclear. The results of the present study showed that the transcription levels of type I IFN were significantly upregulated by a UL24-deleted PRV strain. Furthermore, IFN-β activation induced by poly(dA:dT) or stimulated by cGAS-STING was inhibited by UL24 overexpression in PK15 cells. Co-immunoprecipitation analysis demonstrated that UL24 interacts with and can degrade interferon regulatory factor 7 (IRF7) through the proteasome pathway in a dose-dependent manner. Together, these results showed that PRV UL24 interacted with IRF7 via the proteasome pathway and antagonized cGAS-STING-mediated activation of IFN-β. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
25. The ubiquitin proteasome pathway in neuropsychiatric disorders
- Author
-
Milan Dean, Solmi Cheon, and Maria H. Chahrour
- Subjects
0301 basic medicine ,Proteasome Pathway ,Brain Diseases ,Proteasome Endopeptidase Complex ,Brain development ,biology ,Autism Spectrum Disorder ,Ubiquitin ,Cognitive Neuroscience ,Mental Disorders ,Protein turnover ,Experimental and Cognitive Psychology ,03 medical and health sciences ,Behavioral Neuroscience ,030104 developmental biology ,Proteasome ,Attention Deficit Disorder with Hyperactivity ,Intellectual Disability ,biology.protein ,Schizophrenia ,Humans ,Neuroscience ,Function (biology) ,Metabolic Networks and Pathways - Abstract
The ubiquitin proteasome system (UPS) is a highly conserved pathway that tightly regulates protein turnover in cells. This process is integral to neuronal development, differentiation, and function. Several members of the UPS are disrupted in neuropsychiatric disorders, highlighting the importance of this pathway in brain development and function. In this review, we discuss some of these pathway members, the molecular processes they regulate, and the potential for targeting the UPS in an effort to develop therapeutic strategies in neuropsychiatric and neurodevelopmental disorders.
- Published
- 2017
26. Understanding the interplay between the proteasome pathway and autophagy in response to dual PI3K/mTOR inhibition in myeloma cells is essential for their effective clinical application
- Author
-
Faith E. Davies, Gareth J. Morgan, Fabio Mirabella, Emma L. Davenport, and Lauren I. Aronson
- Subjects
Proteasome Pathway ,Proteasome Endopeptidase Complex ,Cancer Research ,TOR Serine-Threonine Kinases ,Autophagy ,Hematology ,Phosphatidylinositol 3-Kinases ,Biology ,medicine.disease ,Cell biology ,Oncology ,medicine ,Humans ,Proteasome endopeptidase complex ,Multiple Myeloma ,Letter to the Editor ,PI3K/AKT/mTOR pathway ,Multiple myeloma ,Phosphoinositide-3 Kinase Inhibitors - Abstract
Understanding the interplay between the proteasome pathway and autophagy in response to dual PI3K/mTOR inhibition in myeloma cells is essential for their effective clinical application
- Published
- 2013
- Full Text
- View/download PDF
27. HuCOP1 contributes to the regulation of DNA repair in keratinocytes
- Author
-
István Németh, Éva Ádám, Márta Széll, Lajos Kemény, Barbara Fazekas, and Michael P. Carty
- Subjects
0301 basic medicine ,Keratinocytes ,congenital, hereditary, and neonatal diseases and abnormalities ,DNA Repair ,DNA repair ,hmuts-alpha ,Ubiquitin-Protein Ligases ,Clinical Biochemistry ,mammalian homolog ,keratinocyte ,medicine.disease_cause ,Genomic Instability ,03 medical and health sciences ,medicine ,Gene silencing ,Humans ,Molecular Biology ,negative regulator ,Cell Line, Transformed ,Cisplatin ,biology ,Cell Biology ,General Medicine ,hucop1 ,Ubiquitin ligase ,photomorphogenesis ,mismatch repair ,tumorigenesis ,030104 developmental biology ,medicine.anatomical_structure ,MutS Homolog 2 Protein ,proteasome pathway ,MSH2 ,biology.protein ,Cancer research ,human-cells ,DNA mismatch repair ,ubiquitin ligase cop1 ,Tumor Suppressor Protein p53 ,Carcinogenesis ,Keratinocyte ,msh2 ,damage ,genome stability ,medicine.drug - Abstract
We have previously demonstrated that the E3 ligase Human Constitutive Photomorphogenic Protein (huCOP1) is expressed in human keratinocytes and negatively regulates p53. The MutS homolog 2 (MSH2) protein plays a central role in DNA MMR mechanism and is implicated in the cellular response to anticancer agents, such as cisplatin. Our aim was to clarify whether huCOP1 plays a role in DNA MMR by affecting MSH2 protein level in human keratinocytes. To define the role of huCOP1 in DNA mismatch repair, we determined whether huCOP1 affects MSH2 abundance. MSH2 protein level was detected by immunocytochemical staining using a keratinocyte cell line in which the expression level of huCOP1 was stably decreased (siCOP1). To investigate whether huCOP1 silencing influences cisplatin-induced cell death, control and siCOP1 keratinocyte cells were treated with increasing concentrations of cisplatin and cell viability was recorded after 48 and 96 h. Stable silencing of huCOP1 in human keratinocytes resulted in a reduced level of MSH2 protein. huCOP1 silencing also sensitized keratinocytes to the interstrand crosslinking inducer cisplatin. Our results indicate that decreased huCOP1 correlates with lower MSH2 levels. These protein level changes lead to increased sensitivity toward cisplatin treatment, implicating that huCOP1 plays a positive role in maintaining genome integrity in human keratinocytes.
- Published
- 2016
28. FO060A NEW TREATMENT OF PROTEASOME PATHWAY INTERVENTION FOR SEVERE COL4A3 MUTATION CAUSED KIDNEY DISEASE
- Author
-
Jianni Huang, Huidi Zhang, Nan Chen, Hong Ren, Yunzi Liu, and Jingyuan Xie
- Subjects
Proteasome Pathway ,Transplantation ,Nephrology ,business.industry ,Intervention (counseling) ,Mutation (genetic algorithm) ,Cancer research ,medicine ,medicine.disease ,business ,Kidney disease - Published
- 2018
- Full Text
- View/download PDF
29. Targeting the proteasome pathway
- Author
-
Sachiko Tsukamoto and Hideyoshi Yokosawa
- Subjects
Proteasome Endopeptidase Complex ,medicine.medical_treatment ,Clinical Biochemistry ,Antineoplastic Agents ,Ubiquitin-Activating Enzymes ,Targeted therapy ,Bortezomib ,Lactones ,Structure-Activity Relationship ,Drug Delivery Systems ,Ubiquitin ,Neoplasms ,Drug Discovery ,medicine ,Animals ,Humans ,Protease Inhibitors ,Pyrroles ,Ubiquitins ,Multiple myeloma ,Pharmacology ,Proteasome Pathway ,Biological Products ,biology ,business.industry ,Drug discovery ,medicine.disease ,Boronic Acids ,Acetylcysteine ,Cell biology ,Proteasome ,Pyrazines ,Proteasome inhibitor ,biology.protein ,Molecular Medicine ,Peptides ,business ,Proteasome Inhibitors ,medicine.drug - Abstract
Background: The ubiquitin–proteasome pathway functions as a main pathway in intracellular protein degradation and plays a vital role in almost all cellular events. Various inhibitors of this pathway have been developed for research purposes. The recent approval of bortezomib (PS-341, Velcade®), a proteasome inhibitor, for the treatment of multiple myeloma has opened the way to the discovery of drugs targeting the proteasome and other components of the ubiquitin–proteasome pathway. Objectives: We review the current understanding of the ubiquitin–proteasome pathway and inhibitors targeting this pathway, including proteasome inhibitors, as candidate drugs for chemical therapy. Methods: Preclinical and clinical data for inhibitors of the proteasome and the ubiquitin–proteasome pathway are discussed. Conclusions: The proteasome and other members in the ubiquitin–proteasome pathway have emerged as novel therapeutic targets.
- Published
- 2009
- Full Text
- View/download PDF
30. The ubiquitin proteasome pathway in neuropsychiatric disorders.
- Author
-
Cheon, Solmi, Dean, Milan, and Chahrour, Maria
- Subjects
- *
NEUROBEHAVIORAL disorders , *NEURAL development , *NEUROLOGICAL disorders , *AUTISM spectrum disorders - Abstract
• The ubiquitin proteasome system (UPS) tightly regulates cellular protein turnover. • UPS function and regulation is essential for normal brain development. • Genes encoding members of the UPS are mutated in neurodevelopmental disorders. • Modulating the UPS presents a therapeutic opportunity for many neurological disorders. The ubiquitin proteasome system (UPS) is a highly conserved pathway that tightly regulates protein turnover in cells. This process is integral to neuronal development, differentiation, and function. Several members of the UPS are disrupted in neuropsychiatric disorders, highlighting the importance of this pathway in brain development and function. In this review, we discuss some of these pathway members, the molecular processes they regulate, and the potential for targeting the UPS in an effort to develop therapeutic strategies in neuropsychiatric and neurodevelopmental disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
31. PU.1 Is Degraded in Differentiation of Erythrocytes Through a Proteasome-Dependent Pathway
- Author
-
Yuko Konishi and Akira Tominaga
- Subjects
Erythrocytes ,Immunoprecipitation ,Cellular differentiation ,Lactacystin ,Biology ,Cell Line ,chemistry.chemical_compound ,Proto-Oncogene Proteins ,Erythrocyte differentiation ,Genetics ,medicine ,Electrophoresis, Gel, Two-Dimensional ,Molecular Biology ,Transcription factor ,Proteasome Pathway ,Hydrolysis ,Cell Differentiation ,Cell Biology ,General Medicine ,Acetylcysteine ,Cell biology ,Biochemistry ,Proteasome ,chemistry ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,Trans-Activators ,Proteasome inhibitor ,medicine.drug - Abstract
The transcription factor PU.1 regulates erythrocyte differentiation. We previously reported that F5-5 erythroblasts differentiate into erythrocytes in response to activin by degrading PU.1, and that inhibiting PU.1- degradation suppresses F5-5 cell differentiation into erythrocytes. These findings suggest that regulating PU.1 degradation is critical for terminal differentiation of erythrocytes. Here, we investigate the mechanism underlying PU.1 degradation during successive differentiation of erythrocytes. Using 2D-MS proteomic analysis, we show that proteasome subunits and proteins required for degradation by proteasomes immunoprecipitate with PU.1 in response to activin. Furthermore, a proteasome inhibitor, lactacystin, partially suppresses differentiation of F5-5 cells into erythrocytes in response to activin, and partially inhibits PU.1 degradation. Our results indicate that degradation of PU.1 necessary for erythrocyte differentiation occurs, in part, through the proteasome pathway.
- Published
- 2006
- Full Text
- View/download PDF
32. Phosphorylation of mCRY2 at Ser557 in the Hypothalamic Suprachiasmatic Nucleus of the Mouse
- Author
-
Yuko Harada, Yoshitaka Fukada, Mihoko Sakai, Nobuhiro Kurabayashi, and Tsuyoshi Hirota
- Subjects
inorganic chemicals ,Proteasome Endopeptidase Complex ,medicine.medical_specialty ,animal structures ,Physiology ,Circadian clock ,Hypothalamus ,macromolecular substances ,Biology ,Glycogen Synthase Kinase 3 ,Mice ,Oscillometry ,Physiology (medical) ,Internal medicine ,Chlorocebus aethiops ,Serine ,medicine ,Animals ,Circadian rhythm ,Phosphorylation ,Glycogen synthase ,Proteasome Pathway ,Glycogen Synthase Kinase 3 beta ,Flavoproteins ,Suprachiasmatic nucleus ,Immunohistochemistry ,Circadian Rhythm ,Cell biology ,Peripheral ,Cryptochromes ,enzymes and coenzymes (carbohydrates) ,Endocrinology ,Liver ,COS Cells ,biology.protein ,bacteria ,Suprachiasmatic Nucleus - Abstract
Cryptochrome1 and 2 play a critical role in the molecular oscillations of the circadian clocks of central and peripheral tissues in mammals. Mouse Cryptochrome2 (mCRY2) is phosphorylated at Ser557 in the liver, in which the Ser557-phosphorylated form accumulates during the night in parallel with mCRY2 protein. Phosphorylation of mCRY2 at Ser557 allows subsequent phosphorylation at Ser553 by glycogen synthase kinase-3beta (GSK-3beta), resulting in efficient degradation of mCRY2 by a proteasome pathway. In the present study, we found that mCRY2 is phosphorylated at Ser557 also in the region of the mouse brain containing the suprachiasmatic nucleus (SCN), the central circadian clock tissue. Daily fluctuation of the Ser557-phosphorylation level in the SCN region suggests an important role of sequential phosphorylation of Ser557 and Ser553 in the rhythmic degradation of mCRY2 in both central and peripheral clocks of mice.
- Published
- 2006
- Full Text
- View/download PDF
33. Protective Effects of Mutant Ubiquitin in Transgenic Mice
- Author
-
M. Beyers, Maria Tsirigotis, John Woulfe, Jan Brun, Douglas A. Gray, Matthew Y. Tang, and M. Zhang
- Subjects
Genetically modified mouse ,Aging ,Proteasome Endopeptidase Complex ,Time Factors ,Transgene ,Mutant ,Mice, Transgenic ,General Biochemistry, Genetics and Molecular Biology ,Body Temperature ,Mice ,History and Philosophy of Science ,Ubiquitin ,Multienzyme Complexes ,In vivo ,Animals ,Proteasome Pathway ,Genetics ,biology ,Life span ,General Neuroscience ,Body Weight ,Cell biology ,Cysteine Endopeptidases ,Retroviridae ,Mutation ,biology.protein ,Cisplatin ,DNA Damage - Abstract
The K48R mutant ubiquitin can exert profound in vivo protective effects against a variety of insults, including agents of direct clinical relevance. The manipulation of the ubiquitin/proteasome pathway has enormous potential for clinical benefit, and it is not unreasonable to expect that such benefits will include diseases of aging.
- Published
- 2004
- Full Text
- View/download PDF
34. Back to the Future with Ubiquitin
- Author
-
Cecile M. Pickart
- Subjects
Proteasome Pathway ,Proteasome Endopeptidase Complex ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,Ubiquitin ,Models, Biological ,General Biochemistry, Genetics and Molecular Biology ,Histones ,Cysteine Endopeptidases ,Phenotype ,Multienzyme Complexes ,biology.protein ,Animals ,Humans ,Proteasome endopeptidase complex ,Lysosomes ,Neuroscience ,Signal Transduction - Abstract
Two papers published in 1984 by the Varshavsky laboratory revealed that the ubiquitin/proteasome pathway is the principal system for degradation of short-lived proteins in mammalian cells, setting the stage for future demonstrations of this pathway's many regulatory roles. This perspective discusses the impact of those papers and highlights some of the subsequent insights that have led to our current appreciation of the breadth of ubiquitin-mediated signaling.
- Published
- 2004
- Full Text
- View/download PDF
35. Linkage Between the Proteasome Pathway and Neurodegenerative Diseases and Aging
- Author
-
Yves Briand, Sophie Vigouroux, and Mariele Briand
- Subjects
Proteasome Pathway ,Aging ,Proteasome Endopeptidase Complex ,medicine.diagnostic_test ,Proteolysis ,Antigen presentation ,Neuroscience (miscellaneous) ,Regulator ,Neurodegenerative Diseases ,Protein aggregation ,Biology ,Adenosine ,Parkin ,Cell biology ,Cellular and Molecular Neuroscience ,Neurology ,Proteasome ,Biochemistry ,medicine ,Animals ,Humans ,Signal Transduction ,medicine.drug - Abstract
During aging, the production of free radicals increases. This can result in damage to protein, the accumulation of which is characteristic of the aging process. This questions the efficacy of proteolytic systems. Among these systems, the proteasome and the adenosine triphosphate-ubiquitin-dependent pathway have been shown to play an important role in the elimination of abnormal proteins. There are two major steps in the ubiquitin-proteasome pathway: the conjugation of a polyubiquitin degradation signal to the substrate and the subsequent degradation of the tagged protein by the 26S proteasome. The 26S proteasome is build-up from the 20S proteasome, which is a cylinder-shaped multimeric complex, and two additional 19S complexes. The 20S proteasome can also bind to 11S regulator and is then implicated in antigen presentation. These regulators confer a high adaptability on proteasome. With advancing age, predisposition to neurodegenerative diseases increases. These diseases are also characterized by protein aggregation. Several findings such as the presence of ubiquinated proteins, usually broken down by proteasomes, and genetic anomalies involving the ubiquitin-proteasome system (parkin, UCH-L1) suggest a link between the ubiquitin-proteasome pathway and the genesis of these diseases.
- Published
- 2004
- Full Text
- View/download PDF
36. [Untitled]
- Author
-
Gi Yeong Huh, Hong-Jo Choi, Su-Yeong Seo, Hak Hee Kim, Young-Hyun Yoo, Heon Lee, J. H. Jeong, and Jeong Man Kim
- Subjects
Pharmacology ,Proteasome Pathway ,Cancer Research ,Sulindac ,Colorectal cancer ,Chemistry ,Biochemistry (medical) ,Clinical Biochemistry ,Lactacystin ,Pharmaceutical Science ,macromolecular substances ,Cell Biology ,medicine.disease ,digestive system diseases ,chemistry.chemical_compound ,Proteasome activity ,Proteasome ,Apoptosis ,medicine ,Proteasome inhibitor ,medicine.drug - Abstract
The present study was conducted to explore the potential role of proteasome pathway in NSAIDs-induced apoptosis. We employed sulindac as a NSAID, and chose the lactacystin for inhibition of proteasome activity. Assessment of apoptosis and proteasome activity assay were undertaken. We demonstrated that sulindac treatment resulted in a decrease of proteasome activity, and that the co-treatment of a proteasome inhibitor lactacystin potentiated the extent of sulindac-induced apoptosis in HT-29 cells by augmentation of the decrease in proteasome activity. Elucidation of the mechanism underlying the regression of colon cancers by combinations of sulindac and lactacystin seems to be an immediate challenge for the near future.
- Published
- 2003
- Full Text
- View/download PDF
37. Ginkgo biloba extract promoted the astrocyte-mediated clearance of intercellular alpha-Syn via autophagy and proteasome pathway
- Author
-
Jun Hua, Gang Hu, Nuo Yin, and Yi Fan
- Subjects
Proteasome Pathway ,biology ,Chemistry ,Ginkgo biloba ,Applied Mathematics ,General Mathematics ,Autophagy ,Alpha (ethology) ,biology.organism_classification ,Cell biology ,medicine.anatomical_structure ,medicine ,Intracellular ,Astrocyte - Published
- 2018
- Full Text
- View/download PDF
38. Multiple lysines combined in HIV-1 Vif determines the responsiveness to CBF-β
- Author
-
Jianzhang Ma and Youwei Ai
- Subjects
Proteasome Endopeptidase Complex ,Viral protein ,viruses ,Biophysics ,Human immunodeficiency virus (HIV) ,HIV Infections ,Vif Protein ,Biology ,medicine.disease_cause ,Biochemistry ,Core Binding Factor beta Subunit ,Cellular protein ,medicine ,vif Gene Products, Human Immunodeficiency Virus ,Humans ,Molecular Biology ,Infectivity ,Proteasome Pathway ,Virulence ,Lysine ,virus diseases ,Cell Biology ,biochemical phenomena, metabolism, and nutrition ,Viral infectivity factor ,Virology ,Recombinant Proteins ,HEK293 Cells ,Amino Acid Substitution ,Host-Pathogen Interactions ,Proteolysis ,HIV-1 ,Mutagenesis, Site-Directed ,Function (biology) - Abstract
The Vif (viral infectivity factor) protein of human immunodeficiency virus type-1 (HIV-1) is critical for HIV-1 infectivity. CBF-β is required for HIV-1 Vif function, as it increases the steady-state level of the HIV-1 Vif protein to promote host restriction factor APOBEC3 degradation. However, the precise mechanism by which CBF-β promotes HIV-1 Vif levels remains unclear. In the present study, we provided evidences that CBF-β promoted steady-state levels of HIV-1 Vif by inhibiting the degradation of HIV-1 Vif through the proteasome pathway. Our results reveal a new mechanism by which a cellular protein supports viral infectivity by inhibiting viral protein degradation.
- Published
- 2014
39. Genetic and epigenetic studies of amyotrophic lateral sclerosis
- Author
-
Teepu Siddique, Guy A. Rouleau, Mark F. Mehler, Michael J. Strong, Peter Leigh, Ammar Al-Chalabi, and Shin Kwak
- Subjects
Epigenomics ,Axonal Transport ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Humans ,Genetic Predisposition to Disease ,Epigenetics ,Amyotrophic lateral sclerosis ,RNA Processing, Post-Transcriptional ,Gene ,Genetic Association Studies ,030304 developmental biology ,Proteasome Pathway ,0303 health sciences ,business.industry ,Amyotrophic Lateral Sclerosis ,medicine.disease ,3. Good health ,Increased risk ,Neurology ,Disease Progression ,Neurology (clinical) ,Age of onset ,business ,Neuroscience ,030217 neurology & neurosurgery ,Modifier Genes - Abstract
The identification of genetic and epigenetic factors that are associated with an increased risk of developing amyotrophic lateral sclerosis (ALS), or that modify the age of onset or rate of progression, requires a multimodal research strategy, facilitated through international collaboration. The discovery of several ALS genes strongly linked to RNA biology, the proteasome pathway, and axonal transport suggest they have an important role in pathogenesis, but the immense complexity of these processes is also apparent. The increasing rate of genetic discoveries brings the hope of designing more targeted and efficacious therapies.
- Published
- 2013
40. 47 Lurbinectedin (PM01183) specifically targets RNA Pol II for degradation via the proteasome pathway in a transcription and TC-NER dependent fashion
- Author
-
J.F. Martínez-Leal, Carlos M. Galmarini, Carmen Cuevas, G. Santamaria, and Luis García-Fernández
- Subjects
Proteasome Pathway ,Cancer Research ,Oncology ,biology ,Chemistry ,Transcription (biology) ,biology.protein ,Lurbinectedin ,RNA polymerase II ,Cell biology - Published
- 2014
- Full Text
- View/download PDF
41. Probing the proteasome pathway
- Author
-
Alfred L. Goldberg
- Subjects
Proteasome Pathway ,Chemistry ,Biomedical Engineering ,Molecular Medicine ,Bioengineering ,Applied Microbiology and Biotechnology ,Biotechnology ,Cell biology - Published
- 2000
- Full Text
- View/download PDF
42. Evidence for the Existence in Arabidopsis thaliana of the Proteasome Proteolytic Pathway: ACTIVATION IN RESPONSE TO CADMIUM
- Author
-
Polge, Cécile, Jaquinod, Michel, Holzer, Frances, Bourguignon, Jacques, Walling, Linda, Brouquisse, Renaud, Laboratoire de physiologie cellulaire végétale (LPCV), Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Université Joseph Fourier - Grenoble 1 (UJF)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Recherche Agronomique (INRA), Laboratoire d'étude de la dynamique des protéomes (LEDyP), Université Joseph Fourier - Grenoble 1 (UJF)-Commissariat à l'énergie atomique et aux énergies alternatives (CEA)-Institut National de la Santé et de la Recherche Médicale (INSERM), Department of Botany and Plant Sciences [Riverside], University of California [Riverside] (UCR), University of California-University of California, Interactions Biotiques et Santé Végétale (IBSV), Institut National de la Recherche Agronomique (INRA)-Université Nice Sophia Antipolis (... - 2019) (UNS), and COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
MESH: Dipeptidyl-Peptidases and Tripeptidyl-Peptidases ,MESH: Enzyme Activation ,Arabidopsis thaliana ,cadmium ,MESH: Cadmium ,MESH: Arabidopsis Proteins ,oxidized protein degradation ,MESH: Aminopeptidases ,proteomics ,MESH: Up-Regulation ,oxidative stress ,MESH: Animals ,MESH: Arabidopsis ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,MESH: Serine Endopeptidases ,MESH: Gene Expression Regulation, Plant ,MESH: Stress, Physiological ,aminopeptidase ,MESH: Proteasome Endopeptidase Complex ,MESH: Gene Expression Regulation, Enzymologic ,protease ,Plant ,heavy metal ,peptidase ,MESH: Plant Leaves ,MESH: Cattle ,proteasome pathway ,MESH: Muramidase ,MESH: Serum Albumin, Bovine - Abstract
International audience; Heavy metals are known to generate reactive oxygen species that lead to the oxidation and fragmentation of proteins, which become toxic when accumulated in the cell. In this study, we investigated the role of the proteasome during cadmium stress in the leaves of Arabidopsis thaliana plants. Using biochemical and proteomics approaches, we present the first evidence of an active proteasome pathway in plants. We identified and characterized the peptidases acting sequentially downstream from the proteasome in animal cells as follows: tripeptidyl-peptidase II, thimet oligopeptidase, and leucine aminopeptidase. We investigated the proteasome proteolytic pathway response in the leaves of 6-week-old A. thaliana plants grown hydroponically for 24, 48, and 144 h in the presence or absence of 50 mum cadmium. The gene expression and proteolytic activity of the proteasome and the different proteases of the pathway were found to be up-regulated in response to cadmium. In an in vitro assay, oxidized bovine serum albumin and lysozyme were more readily degraded in the presence of 20 S proteasome and tripeptidyl-peptidase II than their nonoxidized form, suggesting that oxidized proteins are preferentially degraded by the Arabidopsis 20 S proteasome pathway. These results show that, in response to cadmium, the 20 S proteasome proteolytic pathway is up-regulated at both RNA and activity levels in Arabidopsis leaves and may play a role in degrading oxidized proteins generated by the stress.
- Published
- 2009
- Full Text
- View/download PDF
43. ChemInform Abstract: Structure of the Complex Between Phosphorylated Substrates and the SCF β-TrCP Ubiquitin Ligase Receptor: A Combined NMR, Molecular Modeling, and Docking Approach
- Author
-
Richard Benarous, Nathalie Evrard-Todeschi, Julien Pons, Josyane Gharbi-Benarous, Jean-Pierre Girault, and Gildas Bertho
- Subjects
Proteasome Pathway ,biology ,Molecular model ,Chemistry ,General Medicine ,computer.file_format ,Protein degradation ,Protein Data Bank ,Ubiquitin ligase ,Docking (molecular) ,biology.protein ,Biophysics ,Phosphorylation ,Receptor ,computer - Abstract
The binding of phosphorylated peptides to the receptor plays a major role in many basic cellular processes in a variety of pathological states. Human β-TrCP is a key component of a recently characterized E3 ubiquitin ligase complex that regulates protein degradation through the ubiquitin-dependent proteasome pathway. Docking studies were carried out to explore the structural requirements for the β-TrCP substrates. Docking studies were performed on the bound conformation of the phosphorylated peptides determined by NMR, whereas the β-TrCP structure was derived by X-ray from Protein Data Bank. After the docking calculation, during which the peptides were conformationally restrained, the complex presented herein was analyzed in terms of ligand-protein interactions and properties of contacting surfaces. The structural requirements for phosphorylated substrates in interaction with β-TrCP were explored and compared with experimental data from TRNOESY and STD NMR results. The analysis revealed that the bend of t...
- Published
- 2009
- Full Text
- View/download PDF
44. Structure of the complex between phosphorylated substrates and the SCF beta-TrCP ubiquitin ligase receptor: a combined NMR, molecular modeling, and docking approach
- Author
-
Julien Pons, Richard Benarous, Gildas Bertho, Nathalie Evrard-Todeschi, Josyane Gharbi-Benarous, and Jean-Pierre Girault
- Subjects
Models, Molecular ,Molecular model ,Macromolecular Substances ,General Chemical Engineering ,Molecular Sequence Data ,Library and Information Sciences ,Biology ,Protein degradation ,In Vitro Techniques ,Crystallography, X-Ray ,Ligands ,Substrate Specificity ,Catalytic Domain ,Consensus Sequence ,Humans ,Computer Simulation ,Amino Acid Sequence ,Phosphorylation ,Receptor ,Nuclear Magnetic Resonance, Biomolecular ,beta Catenin ,Proteasome Pathway ,SKP Cullin F-Box Protein Ligases ,Sequence Homology, Amino Acid ,General Chemistry ,computer.file_format ,Protein Data Bank ,Computer Science Applications ,Ubiquitin ligase ,Biochemistry ,Docking (molecular) ,Biophysics ,biology.protein ,Peptides ,computer - Abstract
The binding of phosphorylated peptides to the receptor plays a major role in many basic cellular processes in a variety of pathological states. Human beta-TrCP is a key component of a recently characterized E3 ubiquitin ligase complex that regulates protein degradation through the ubiquitin-dependent proteasome pathway. Docking studies were carried out to explore the structural requirements for the beta-TrCP substrates. Docking studies were performed on the bound conformation of the phosphorylated peptides determined by NMR, whereas the beta-TrCP structure was derived by X-ray from Protein Data Bank. After the docking calculation, during which the peptides were conformationally restrained, the complex presented herein was analyzed in terms of ligand-protein interactions and properties of contacting surfaces. The structural requirements for phosphorylated substrates in interaction with beta-TrCP were explored and compared with experimental data from TRNOESY and STD NMR results. The analysis revealed that the bend of the peptide structures, which is indispensable for beta-TrCP recognition, aligns two charged phosphate groups and a central hydrophobic group in a favorable arrangement that leads to the burial of the peptide surface in the binding cleft upon complexation. Through docking simulations, we have identified different specific binding pockets of beta-TrCP according to the ligand in interaction. These data should be valuable in the rational design of a ligand to be used in therapeutic approaches.
- Published
- 2008
45. Identification of novel mouse Delta1 target genes
- Author
-
Hutterer, Christine, Adamski, Jerzy (Prof. Dr.), Wurst, Wolfgang (Prof. Dr.), and Gierl, Alfons (Prof. Dr.)
- Subjects
Biowissenschaften, Biologie ,ddc:570 ,Notch ,Dll1 ,Delta-like 1 ,Delta1 ,Dll3 ,Jag1 ,Ifitm1 ,Mil-2 ,Smarcc1 ,Srg3 ,SWI/SNF ,Nsf ,14-3-3 ,Csk ,Somitogenesis ,Microarray ,Transcriptome ,Proteome ,Proteasome pathway ,Intracellular trafficking ,Somitogenese ,Transkriptom ,Proteom ,Proteasome Pathway ,Intrazellulärer Transport - Abstract
The Delta/Notch signal transduction pathway is an evolutionary conserved pathway involved in many diverse developmental processes. These include neurogenesis, somitogenesis, left-right development, pancreatic development and development of the sensory hair in the inner ear. Numerous genes have been identified in the last years that are part of the signal transduction pathway or can influence the pathway in a way. A model to explain the process of lateral inhibition, one of the main features of the pathway, has been established. However, it is not clear how the known genes involved in Delta/Notch signalling can account for such diverse processes. In order to get new insights into the regulation of Delta/Notch signalling and to identify targets of Dll1 the transcriptome and the proteome of E10.5 wildtype and Dll1-/- embryos was analysed using DNA-chip technology and 2D-gelelectrophoresis combined with mass spectrometry. In the transcriptome analysis 22 up- and 30 downregulated transcripts were identified. In the proteome analysis 13 proteins were up- and 37 proteins were downregulated at E10.5. Further methods to verify the data on transcriptomics side included whole mount in situ hybridisation and real-time PCR and semi-quantitative immunoblotting on the proteomics side. The most promising candidate out of this analyses is the gene Ifitm1 which shows expression in regions where Delta/Notch signalling is known to occur, namely the presomitic mesoderm and the latest formed somites. The expression was strongly reduced in Dll1, Dll3 and Jag1 mutants which indicates a direct connection between Dll1 or Delta/Notch signalling respectively, and Ifitm1. Furthermore Ifitm1 was only recently discovered as marker for primordial germ cells and has no assigned function so far. To assess the exact function of Ifitm1 in the mouse a classical knockout targeting construct has been designed and prepared. The knockout animals will provide insights into the relevance of Ifitm1 for somitogenesis and other developmental processes. Further genes have been identified which show expression patterns indicative of Delta/Notch signalling and which also were altered in Dll1 mutant mouse embryos, and some also in Dll3 and Jag1 embryos. These genes are Csk, Ddx6, Nes, Sema5b and Smarcc1. On the protein level a number of interesting proteins have been identified. These include six subunits of the 26S proteasome, four translation initiation or elongation factors, proteins involved in cell signalling such as two 14-3-3 proteins and proteins involved in intracellular transport. Taken the data from the transcriptome and proteome analyses together it seems to become clear that in the Dll1 mutant many cellular processes might be altered. Regulation on the genomic level seems to be disturbed at the process of chromatin remodelling, regulation on post-transcriptional levels seems to be disturbed at the process of translation initiation and elongation as well as at the process of protein degradation through the proteasome. Intracellular trafficking seems to be altered on the level of phosphorylation and dephosphorylation through protein phosphatase 2A (PP2A) and C-src tyrosine kinase (Csk), as well as through the 14-3-3 proteins. Further aspects of intracellular trafficking involve the cytoskeleton. Nestin and g-tubulin, two components of the cytoskeleton were deregulated in the Dll1 mutant. Furthermore intracellular membrane trafficking seems to be disrupted through the downregulation of the SNARE protein Nsf and its interaction partner Munc18-3. It also seemed to turn out that developmental processes are altered in the mutants. Four transcription factors have been identified which have not been brought into context with Delta/Notch signalling before. It is not clear yet if they might play roles on the side of the signalling (Delta expressing) or the receiving (Notch expressing) cell. Neural crest cell migration also seems to be altered since Sema5b a gene involved in axonal guidance was downregulated in the Dll1 mutant embryos. Using a combination of transcriptomics and the proteomics approaches it could be shown that RNA- and protein expression profiling are both versatile and powerful approaches which complement each other. Thus it was possible to gain new insights into regulatory processes which would not have been possible with one approach alone. Der Delta/Notch Signaltransduktionsweg ist eine an vielfältigen Entwicklungsprozessen beteiligte, evolutionär konservierte Signalkaskade. Bisher wurde die Wirkungsweise während der Neurogenese, Somitogenese, links-rechts Achsenbildung, Pakreasentwicklung und der Entwicklung der sensorischen Härchen im Innenohr beobachtet. In den letzten Jahren wurde eine Anzahle von Genen identifiziert, die direkt an der Signaltransduktion beteiligt sind, oder die den Wechselweg auf verschiedene Weise beeinflussen. Für den wichtigen Entwicklungsprozess der lateralen Inhibition, der von Delta/Notch Signaltransduktion gesteuert wird, wurde ein Modell etabliert. Es bleibt jedoch unklar, wie die begrenzte Anzahl an bisher identifizierten Genen eine solche Vielfalt an Entwicklungsprozessen steuern kann. Um neue Einblicke in die Regulation der Delta/Notch Signaltransduktionskaskade zu gewinnen und um Zielgene von Dll1 zu identifizieren wurde das Transkriptom, sowie das Proteom von 10.5 dpc Wildtyp und Dll1 mutanten Mausembryonen untersucht. Dazu wurde die DNA-Chip Technologie und 2D-Gelelektrophorese kombiniert mit Massenspektrometrie durchgeführt. Durch die Transkriptomanalyse wurden in der Dll1 Mutante 22 hoch- und 30 herunter regulierte Gene identifiziert. Die Proteomanalyse lieferte 13 hoch- und 37 herunter regulierte Proteine am Tag 10.5 der Embryonalentwicklung. Zur Bestätigung und weiteren Analyse der identifizierten Kandidaten wurde Real-Time PCR und Ganzkörper in situ Hybridisierung auf Nukleinsäure-Ebene und semi-quantitatives Immunoblotting auf Protein-Ebene durchgeführt. Der aussichtsreichste Kandidat der Analysen ist das Gen Ifitm1. Es ist in Regionen des Embryos exprimiert von denen man weiß, dass dort Delta/Notch Signalübertragung stattfindet, wie beispielsweise im präsomitischen Mesoderm und den jüngsten Somiten. In Dll1, Dll3 und Jag1 mutanten Mausembryonen war die Expression in diesen Regionen deutlich reduziert, was auf eine direkte Verbindung zwischen Dll1, bzw. Delta/Notch Signaltransduktion und Ifitm1 hinweist. Ifitm1 wurde erst kürzlich als Markergen für primordiale Keimzellen identifiziert, eine genaue Funktion ist bisher nicht bekannt. Um die Funktion von Ifitm1 detailliert untersuchen zu können wurde ein klassisches Targeting Konstrukt entworfen und hergestellt. Mit Hilfe der Knock-out Tiere werden neue Einblicke in die Bedeutung von Ifitm1 für die Somitogenese und andere Entwicklungsprozesse gewonnen werden. Es wurden weitere Gene gefunden, die ein bezüglich Delta/Notch Signaltransduktion bemerkenswertes Expressionsmuster zeigen, und deren Expression in Dll1 Mutanten, sowie auch in Dll3 und Jag1 Mutanten verändert war. Diese Gene sind Csk, Ddx6, Nes, Sema5b and Smarcc1. Auch auf Protein-Ebene wurde eine Reihe interessanter Proteine identifiziert. Dazu gehören beispielsweise sechs Untereinheiten des 26S Proteasoms, vier „translation initiation“ bzw. „elongation“ Faktoren, Proteine der Signalisierungskaskade der Zellen, wie zwei 14-3-3 Proteine und Proteine, die in den intrazellulären Transport involviert sind. Betrachtet man die Daten der Transkriptom- und Proteomanalyse gemeinsam so scheint klar zu werden, dass in der Dll1 Mutante eine Anzahl an zellulären Prozessen beeinträchtigt sein könnte. Auf genomischer Ebene scheint die Regulation der Chromatin Modellierung verändert zu sein. Die post-transkriptionelle Regulation scheint auf der Ebene der Initiierung sowie der Elongation der Transkription ebenso gestört zu sein, wie auch bei dem Prozess der Protein Degradation am Proteasom. Intrazelluläre und regulatorische Transportmechanismen scheinen auf der Ebene der Phosphorylierung und Dephosphorylierung durch Protein Phosphatase 2A (PP2A) und C-src Tyrosin Kinase (Csk) und auch durch 14-3-3 Proteine verändert zu sein. Intrazellulärer Transport hängt weiterhin vom Cytoskelett ab. Zwei Komponenten des Cytoskeletts, Nestin und g-Tubulin, sind reguliert in der Dll1 Mutante. Durch verminderte Expression des SNARE Proteins Nsf, wie auch dessen Interaktionspartner Munc18-3, scheinen auch Transportmechanismen, die mit Hilfe der Zellmembran ablaufen, beeinträchtigt zu sein. Weiterhin scheinen auch Entwicklungsprozesse in der Mutante betroffen zu sein. Beispielsweise wurden vier Transkriptionsfaktoren identifiziert, die noch nie mit der Delta/Notch Signaltransduktion in Zusammenhang gebracht wurden. Im Moment bleibt offen, ob diese Transkriptionsfaktoren auf der Seite der Delta exprimierenden, signalgebenden Zelle, oder auf der Seite der Notch exprimierenden, signalempfangenden Zelle eine Rolle spielen. Eine mögliche Beeinträchtigung der Wanderung der Neuralleistenzellen könnte durch die verminderte Expression von Sema5b angezeigt werden. Sema5b ist in den Prozess der Wegleitung der Neuronen involviert. Mit Hilfe einer Kombination aus Analysen des Transkriptoms und des Proteoms konnte gezeigt werden, dass RNA- und Protein-Expression Profiling zwei vielseitige und leistungsstarke Ansätze sind, die einander komplementieren. Es war auf diese Weise möglich neue Einblicke in regulatorische Mechanismen zu erhalten, die man mit einer Methode allein nicht hätte bekommen können.
- Published
- 2008
46. Biochemical Functions of Ubiquitin and Ubiquitin-like Protein Conjugation
- Author
-
Mark Hochstrasser
- Subjects
Proteasome Pathway ,biology ,Ubiquitin ,Chemistry ,biology.protein ,SUMO protein ,Ubiquitin-conjugating enzyme ,Ubiquitin ligase ,Cell biology - Published
- 2008
- Full Text
- View/download PDF
47. Identification of novel mouse Delta1 target genes: Combination of transcriptomics and proteomics
- Author
-
Hutterer, C.
- Subjects
Notch ,Dll1 ,Delta-like 1 ,Delta1 ,Dll3 ,Jag1 ,Ifitm1 ,Mil-2 ,Smarcc1 ,Srg3 ,SWI/SNF ,Nsf ,14-3-3 ,Csk ,Somitogenesis ,Microarray ,Transcriptome ,Proteome ,Proteasome pathway ,Intracellular trafficking ,Ifitm - Abstract
The Delta/Notch signal transduction pathway is an evolutionary conserved pathway involved in many diverse developmental processes. These include neurogenesis, somitogenesis, left-right development, pancreatic development and development of the sensory hair in the inner ear. Numerous genes have been identified in the last years that are part of the signal transduction pathway or can influence the pathway in a way. A model to explain the process of lateral inhibition, one of the main features of the pathway, has been established. However, it is not clear how the known genes involved in Delta/Notch signalling can account for such diverse processes. In order to get new insights into the regulation of Delta/Notch signalling and to identify targets of Dll1 the transcriptome and the proteome of E10.5 wildtype and Dll1-/- embryos was analysed using DNA-chip technology and 2D-gelelectrophoresis combined with mass spectrometry. In the transcriptome analysis 22 up- and 30 downregulated transcripts were identified. In the proteome analysis 13 proteins were up- and 37 proteins were downregulated at E10.5. Further methods to verify the data on transcriptomics side included whole mount in situ hybridisation and real-time PCR and semi-quantitative immunoblotting on the proteomics side. The most promising candidate out of this analyses is the gene Ifitm1 which shows expression in regions where Delta/Notch signalling is known to occur, namely the presomitic mesoderm and the latest formed somites. The expression was strongly reduced in Dll1, Dll3 and Jag1 mutants which indicates a direct connection between Dll1 or Delta/Notch signalling respectively, and Ifitm1. Furthermore Ifitm1 was only recently discovered as marker for primordial germ cells and has no assigned function so far. To assess the exact function of Ifitm1 in the mouse a classical knockout targeting construct has been designed and prepared. The knockout animals will provide insights into the relevance of Ifitm1 for somitogenesis and other developmental processes. Further genes have been identified which show expression patterns indicative of Delta/Notch signalling and which also were altered in Dll1 mutant mouse embryos, and some also in Dll3 and Jag1 embryos. These genes are Csk, Ddx6, Nes, Sema5b and Smarcc1. On the protein level a number of interesting proteins have been identified. These include six subunits of the 26S proteasome, four translation initiation or elongation factors, proteins involved in cell signalling such as two 14-3-3 proteins and proteins involved in intracellular transport. Taken the data from the transcriptome and proteome analyses together it seems to become clear that in the Dll1 mutant many cellular processes might be altered. Regulation on the genomic level seems to be disturbed at the process of chromatin remodelling, regulation on post-transcriptional levels seems to be disturbed at the process of translation initiation and elongation as well as at the process of protein degradation through the proteasome. Intracellular trafficking seems to be altered on the level of phosphorylation and dephosphorylation through protein phosphatase 2A (PP2A) and C-src tyrosine kinase (Csk), as well as through the 14-3-3 proteins. Further aspects of intracellular trafficking involve the cytoskeleton. Nestin and g-tubulin, two components of the cytoskeleton were deregulated in the Dll1 mutant. Furthermore intracellular membrane trafficking seems to be disrupted through the downregulation of the SNARE protein Nsf and its interaction partner Munc18-3. It also seemed to turn out that developmental processes are altered in the mutants. Four transcription factors have been identified which have not been brought into context with Delta/Notch signalling before. It is not clear yet if they might play roles on the side of the signalling (Delta expressing) or the receiving (Notch expressing) cell. Neural crest cell migration also seems to be altered since Sema5b a gene involved in axonal guidance was downregulated in the Dll1 mutant embryos. Using a combination of transcriptomics and the proteomics approaches it could be shown that RNA- and protein expression profiling are both versatile and powerful approaches which complement each other. Thus it was possible to gain new insights into regulatory processes which would not have been possible with one approach alone.
- Published
- 2005
48. Rings, chains and ladders: ubiquitin goes to work in the neuron
- Author
-
Jennifer A Johnston and Kiran Madura
- Subjects
Proteasome Endopeptidase Complex ,Protein degradation ,Biology ,Ubiquitin ,Multienzyme Complexes ,medicine ,Protein biosynthesis ,Animals ,Humans ,Proteasome Pathway ,Neurons ,Neuronal Plasticity ,General Neuroscience ,Neurodegenerative Diseases ,Neuronal Pathfinding ,Endocytosis ,Cell biology ,Cysteine Endopeptidases ,medicine.anatomical_structure ,Proteasome ,Synaptic plasticity ,Synapses ,biology.protein ,Neuron ,Neuroscience ,Signal Transduction - Abstract
Our understanding of neuronal cell biology in the last 10 years has exploded. In parallel, our grasp of basic cellular processes, such as protein synthesis and protein degradation has also grown exponentially. In this review, we provide an in-depth background to details of current knowledge of the Ub/proteasome pathway. We also provide examples of recent experiments in neurobiology that suggest a central role for targeted protein degradation by the Ub/proteasome pathway to ensure proper neuronal function. From the examples provided, it is clear the activity of the proteasome is required for neuronal pathfinding during development, regulation of synaptic branching and number, and synaptic plasticity. We conclude with a discussion of how defects in proteasome pathway function may lead to neuronal dysfunction, with specific emphasis on diseases characterized by the accumulation of ubiquitin (Ub)-positive inclusions. Our goal is to excite the expert neurobiologist to the myriad ways that specific neuronal functions could be regulated (or dysregulated) by mechanisms involving the Ub/proteasome pathway.
- Published
- 2004
49. Inhibition of Fatty Acid Synthase in Prostate Cancer by Orlistat, a Novel Therapeutic
- Author
-
WAKE FOREST UNIV WINSTON-SALEM NC SCHOOL OF HEALTH SCIENCES, Kridel, Steven J., WAKE FOREST UNIV WINSTON-SALEM NC SCHOOL OF HEALTH SCIENCES, and Kridel, Steven J.
- Abstract
The basic premise of this proposal was based on two important findings. The first was the discovery that fatty acid synthase (FAS) is overexpressed in prostate cancer as well as in many other cancers. FAS is the enzyme that catalyzes the synthesis of fatty acid from the precursors acetyl-CoA and malonyl-CoA. The fatty acids are then utilized for subsequent phospholipid synthesis and membrane biogenesis. A body of literature has demonstrated that tumor cells are addicted to FAS derived fatty acids as inhibition of FAS activity induces cell death in tumor cells. The second was the initial discovery of the FDA-approved drug Orlistat as an inhibitor of FAS. Orlistat targets the thioesterase domain of FAS and induces cell death specifically in prostate tumor cells and inhibits the growth of prostate tumor xenografts in mice. Based on these discoveries, three specific aims were proposed. They were as follows: (1) to determine the cellular consequences of FAS inhibition by Orlistat, (2) to analyze the molecular basis for FAS inhibition by Orlistat, and (3) to select and characterize novel FAS inhibitor scaffolds using peptide phage display. The overall goal of this project was to understand the anti-tumor effects of FAS inhibitors. The authors have followed up the endoplasmic reticulum (ER) stress response. Moreover, they have identified crosstalk between the FAS and proteasome pathway that, when perturbed, enhances ER stress signaling. In addition, preliminary data suggests that the ER stress and autophagy pathways may be acting in concert when FAS activity is reduced in tumor cells. In another line of investigation, they solved the first crystal structure of a FAS domain bound to ligand as well as the structure of the thioesterase domain of FAS bound to Orlistat, in two states. Together, these data provide a framework, or blueprint, for the design of novel FAS inhibitors and an understanding of their anti-tumor mechanisms and their future translation into the clinic., The original document contains color images.
- Published
- 2007
50. Functional overload attenuates plantaris atrophy in tumor-bearing rats
- Author
-
Otis, Jeffrey S., Lees, Simon J., Williams, Jay H., Otis, Jeffrey S., Lees, Simon J., and Williams, Jay H.
- Abstract
Background Late stage cancer malignancies may result in severe skeletal muscle wasting, fatigue and reduced quality of life. Resistance training may attenuate these derangements in cancer patients, but how this hypertrophic response relates to normal muscle adaptations in healthy subjects is unknown. Here, we determined the effect of resistance training on muscle mass and myosin heavy chain (MHC) isoform composition in plantaris muscles from tumor-bearing (TB) rats. Methods Age- and gender-matched Buffalo rats were used for all studies (n = 6/group). Suspensions of Morris Hepatoma MH7777 cells or normal saline were injected subcutaneously into the dorsum. Six weeks after cell implantation, muscles from TB rats were harvested, weighed and processed for ATP-independent proteasome activity assays. Once tumor-induced atrophy had been established, subgroups of TB rats underwent unilateral, functional overload (FO). Healthy, sham-operated rats served as controls. After six weeks, the extent of plantaris hypertrophy was calculated and MHC isoform compositions were determined by gel electrophoresis. Results Six weeks of tumor growth reduced body mass and the relative masses of gastrocnemius, plantaris, tibialis anterior, extensor digitorum longus, and diaphragm muscles (p ≤ 0.05). Percent reductions in body mass had a strong, negative correlation to final tumor size (r = -0.78). ATP-independent proteasome activity was increased in plantaris muscles from TB rats (p ≤ 0.05). In healthy rats, functional overload (FO) increased plantaris mass ~44% compared to the contralateral control muscle, and increased the relative percentage of MHC type I and decreased the relative percentage of MHC type IIb compared to the sham-operated controls (p ≤ 0.05). Importantly, plantaris mass was increased ~24% in TB-FO rats and adaptations to MHC isoform composition were consistent with normal, resistance-trained muscles. Conclusion Despite significant skeletal muscle derangements due to cancer
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.