Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Exp Gerontol ; 164: 111831, 2022 07.
Article in English | MEDLINE | ID: mdl-35525396

ABSTRACT

AIMS: This study aimed to evaluate the impact of a 12-week calorie-restricted diet and recreational sports training on gene expressions IL-15, ATROGIN-1 and MURF-1 in skeletal muscle of T2D patients. METHODS: Older adults with T2D (n = 39, 60 ± 6.0 years, BMI 33.5 ± 0.6 kg/m2) were randomly allocated to Diet+Soccer (DS), Diet+Running (DR) or Diet (D). The training sessions were moderate-to-high-intensity and performed 3 × 40 min/week for 12-weeks. Gene expression from vastus lateralis muscle obtained by qRT-PCR, dual-energy X-ray and fasting blood testing measurements were performed before and after 12-weeks. Statistical analysis adopted were two-way ANOVA and Paired t-test for gene expression, and RM-ANOVA test for the remainder variables. RESULTS: Total body weight was reduced in ~4 kg representing body fat mass in all groups after 12-weeks (P < 0.05). HbA1c values decreased in all groups post-intervention. Lipids profile improved in the training groups (P < 0.05) after 12-weeks. ATROGIN-1 and MURF-1 mRNA reduced in the DS (1.084 ± 0.14 vs. 0.754 ± 1.14 and 1.175 ± 0.34 vs. 0.693 ± 0.12, respectively; P < 0.05), while IL-15 mRNA increased in the DR (1.056 ± 0.12 vs. 1.308 ± 0.13; P < 0.05) after 12-weeks intervention. CONCLUSION: Recreational training with a moderate calorie-restricted diet can downregulates the expression of atrophy-associated myokines and increases the expression of anti-inflammatory gene IL-15.


Subject(s)
Caloric Restriction , Diabetes Mellitus, Type 2 , Exercise , Muscle, Skeletal , Aged , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/therapy , Exercise/physiology , Gene Expression , Humans , Interleukin-15/biosynthesis , Interleukin-15/genetics , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , SKP Cullin F-Box Protein Ligases/biosynthesis , SKP Cullin F-Box Protein Ligases/genetics , Tripartite Motif Proteins/biosynthesis , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
2.
Biochem Biophys Res Commun ; 550: 134-141, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33691199

ABSTRACT

Tripartite motif protein 32 (TRIM32), an E3 ubiquitin ligase, has been reported to participate in many human cancers. However, the underlying role of TRIM32 in glioma remains largely unknown. Here, we aimed to explore the function of TRIM32 in glioma cells and the clinical implications and found that TRIM32 was upregulated in glioma tissues. Consistently, overexpression of TRIM32 promoted glioma U87 and U251 cell proliferation and conferred cell resistance to temozolomide (TMZ). Conversely, knockdown of TRIM32 inhibited glioma cells proliferation in vitro and in vivo and sensitized glioma cells to the treatment of TMZ in a p53-dependent and -independent manner. Mechanistically, knockdown of TRIM32 induced apoptosis of U87 an U251 cells. In addition, TRIM32 interacted with the antiapoptotic proteins BCL-xL and BCL-w, which antagonized the inhibitory effect of TRIM32 knockdown in U87 cells. Together, our study uncovered the role of TRIM32 in glioma and TRIM32 may be a potential therapeutic target for gliomas.


Subject(s)
Cell Proliferation , Drug Resistance, Neoplasm , Glioma/drug therapy , Glioma/pathology , Temozolomide/therapeutic use , Transcription Factors/deficiency , Tripartite Motif Proteins/deficiency , Tumor Suppressor Protein p53 , Ubiquitin-Protein Ligases/deficiency , Animals , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioma/genetics , Humans , Mice , Molecular Targeted Therapy , Neoplasm Grading , Temozolomide/pharmacology , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/metabolism , Tripartite Motif Proteins/biosynthesis , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Up-Regulation , Xenograft Model Antitumor Assays
3.
Kaohsiung J Med Sci ; 37(7): 547-561, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33728810

ABSTRACT

Circular RNAs (circRNAs) appear to be significant modulators in various physiological processes. Recently, it is found that circRNA_101996 exerts important roles in various cancers. Our previous studies showed that circRNA_101996 promoted cervical cancer growth and metastasis by regulating miR-8075/TPX2. However, the potential regulatory role of circRNA_101996 in cervical cancer still needs further investigation. Our results in this study suggested that circRNA_101996 was over-expressed in cervical cancer patients. circRNA_101996 up-regulation remarkably assisted cell proliferation, cell cycle progression, and cell migration in cervical cancer, while circRNA_101996 knockdown exerted the inverse effects. The molecular investigations indicated that circRNA_101996 could increase the expression level of miR-1236-3p, tripartite motif-containing 37 (TRIM37), through binding to miR-1236-3p and reducing its expression. Moreover, in vivo results demonstrated that circRNA_101996 shRNA can function as a tumor suppressor through down-regulating TRIM37 in cervical cancer. In conclusion, our data indicated that circRNA_101996/miR-1236-3p/TRIM37 axis accelerated cervical cancer development, providing novel insights into cervical cancer diagnosis and treatment.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , RNA, Circular/genetics , Tripartite Motif Proteins/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Uterine Cervical Neoplasms/genetics , 3' Untranslated Regions , Adult , Aged , Cell Cycle , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , Epithelium/metabolism , Female , Humans , Lymphatic Metastasis , Middle Aged , Neoplasm Metastasis , RNA/metabolism , RNA, Small Interfering/metabolism , Uterine Cervical Neoplasms/metabolism
4.
Am J Physiol Endocrinol Metab ; 320(1): E150-E159, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33284091

ABSTRACT

Sepsis remains a leading cause of mortality in critically ill patients. Muscle wasting is a major complication of sepsis and negatively affects clinical outcomes. Despite intense investigation for many years, the molecular mechanisms underlying sepsis-related muscle wasting are not fully understood. In addition, a potential role of muscle wasting in disease development of sepsis has not been studied. Myostatin is a myokine that downregulates skeletal muscle mass. We studied the effects of myostatin deficiency on muscle wasting and other clinically relevant outcomes, including mortality and bacterial clearance, in mice. Myostatin deficiency prevented muscle atrophy along with inhibition of increases in muscle-specific RING finger protein 1 (MuRF-1) and atrogin-1 expression and phosphorylation of signal transducer and activator of transcription protein 3 (STAT3; major players of muscle wasting) in septic mice. Moreover, myostatin deficiency improved survival and bacterial clearance of septic mice. Sepsis-induced liver dysfunction, acute kidney injury, and neutrophil infiltration into the liver and kidney were consistently mitigated by myostatin deficiency, as indicated by plasma concentrations of aspartate aminotransferase (AST), alanine aminotransferase (ALT), and neutrophil gelatinase-associated lipocalin (NGAL) and myeloperoxidase activity in the organs. Myostatin deficiency also inhibited sepsis-induced increases in plasma high-mobility group protein B1 (HMGB1) and macrophage inhibitory cytokine (MIC)-1/growth differentiation factor (GDF)-15 concentrations. These results indicate that myostatin plays an important role not only in muscle wasting but also in other clinically relevant outcomes in septic mice. Furthermore, our data raise the possibility that muscle wasting may not be simply a complication, but myostatin-mediated muscle cachexia and related changes in muscle may actually drive the development of sepsis as well.NEW & NOTEWORTHY Muscle wasting is a major complication of sepsis, but its role in the disease development is not known. Myostatin deficiency improved bacterial clearance and survival and mitigated damage in the liver and kidney in septic mice, which paralleled prevention of muscle wasting. These results raise the possibility that muscle wasting may not simply be a complication of sepsis, but myostatin-mediated cachexic changes may have a role in impaired bacterial clearance and mortality in septic mice.


Subject(s)
Muscular Atrophy/genetics , Myostatin/deficiency , Myostatin/genetics , Sepsis/genetics , Acute Kidney Injury/genetics , Animals , Cachexia/genetics , Cachexia/prevention & control , Lipocalin-2/blood , Liver Diseases/etiology , Liver Diseases/genetics , Liver Function Tests , Male , Mice , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Muscular Atrophy/prevention & control , Neutrophil Infiltration/genetics , Phosphorylation , STAT3 Transcription Factor/biosynthesis , STAT3 Transcription Factor/genetics , Sepsis/microbiology , Sepsis/mortality , Survival Analysis , Tripartite Motif Proteins/biosynthesis , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
5.
Respir Res ; 21(1): 132, 2020 May 29.
Article in English | MEDLINE | ID: mdl-32471489

ABSTRACT

BACKGROUND: Chronic tissue injury was shown to induce progressive scarring in fibrotic diseases such as idiopathic pulmonary fibrosis (IPF), while an array of repair/regeneration and stress responses come to equilibrium to determine the outcome of injury at the organ level. In the lung, type I alveolar epithelial (ATI) cells constitute the epithelial barrier, while type II alveolar epithelial (ATII) cells play a pivotal role in regenerating the injured distal lungs. It had been demonstrated that eukaryotic cells possess repair machinery that can quickly patch the damaged plasma membrane after injury, and our previous studies discovered the membrane-mending role of Tripartite motif containing 72 (TRIM72) that expresses in a limited number of tissues including the lung. Nevertheless, the role of alveolar epithelial cell (AEC) repair in the pathogenesis of IPF has not been examined yet. METHOD: In this study, we tested the specific roles of TRIM72 in the repair of ATII cells and the development of lung fibrosis. The role of membrane repair was accessed by saponin assay on isolated primary ATII cells and rat ATII cell line. The anti-fibrotic potential of TRIM72 was tested with bleomycin-treated transgenic mice. RESULTS: We showed that TRIM72 was upregulated following various injuries and in human IPF lungs. However, TRIM72 expression in ATII cells of the IPF lungs had aberrant subcellular localization. In vitro studies showed that TRIM72 repairs membrane injury of immortalized and primary ATIIs, leading to inhibition of stress-induced p53 activation and reduction in cell apoptosis. In vivo studies demonstrated that TRIM72 protects the integrity of the alveolar epithelial layer and reduces lung fibrosis. CONCLUSION: Our results suggest that TRIM72 protects injured lungs and ameliorates fibrosis through promoting post-injury repair of AECs.


Subject(s)
Alveolar Epithelial Cells/metabolism , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/prevention & control , Lung/metabolism , Tripartite Motif Proteins/biosynthesis , Alveolar Epithelial Cells/drug effects , Animals , Bleomycin/toxicity , Female , HEK293 Cells , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Lung/drug effects , Male , Mice , Mice, 129 Strain , Mice, Knockout , Recombinant Proteins/biosynthesis
6.
J Atheroscler Thromb ; 27(10): 1039-1052, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32173683

ABSTRACT

AIM: Myostatin (Mstn) has been described as a trigger for the progression of atherosclerosis. In this study, we evaluated the role of Mstn in arterial remodeling in patients with end-stage renal disease (ESRD). METHODS: Vascular specimens were collected from 16 ESRD patients (56.4±7.9 years) undergoing renal transplant (recipients) and 15 deceased kidney non-uremic donors (55.4±12.1 years). We studied gene and protein expression of Mstn, ubiquitin ligases, Atrogin-1, and muscle ring finger protein-1 (MuRF-1), inflammatory marker CCL2, cytoskeleton components, and Klotho by reverse transcription-polymerase chain reaction (RT-PCR) and immunohistochemistry. Moreover, we assessed vascular calcification and collagen deposition. Finally, we studied the effects of recombinant Mstn on rat vascular smooth muscle cells (VSMCs, A7r5) and evaluated the effects of uremic serum (US) on primary human VSMCs. RESULTS: Myostatin mRNA was upregulated in the arterial vascular wall of recipients compared with donors (~15- folds, p<0.05). This response was accompanied by the upregulation of gene expression of Atrogin-1 and MuRF-1 (+2.5- and +10-fold) and CCL2 (+3-fold). Conversely, we found downregulation of protein expression of Smoothelin, α-smooth muscle actin (α-SMA), vimentin, and Klotho (-85%, -50%, -70%, and -80%, respectively; p<0.05) and gene expression of vimentin and Klotho. Exposition of A7r5 to Mstn induced a time-dependent SMAD 2/SMAD 3 phosphorylation and expression of collagen-1 and transforming growth factor ß (TGFß) mRNA, while US induced overexpression of Mstn and Atrogin-1 and downregulation of Smoothelin and Klotho. CONCLUSIONS: Our data suggest that uremia might induce vascular Mstn gene expression together with a complex pathway of molecular and structural changes in the vascular wall. Myostatin, in turn, can translate the metabolic alterations of uremia into profibrotic and stiffness inducing signals.


Subject(s)
Arteries/pathology , Endothelium, Vascular/pathology , Kidney Failure, Chronic/metabolism , Myostatin/metabolism , Adolescent , Adult , Aged , Animals , Chemokine CCL2/biosynthesis , Collagen/metabolism , Cytoskeleton/metabolism , Female , Gene Expression Regulation , Glucuronidase/biosynthesis , Humans , Inflammation , Kidney Failure, Chronic/physiopathology , Kidney Transplantation , Klotho Proteins , Male , Middle Aged , Muscle Proteins/biosynthesis , Muscle, Smooth, Vascular/metabolism , Rats , SKP Cullin F-Box Protein Ligases/biosynthesis , Tripartite Motif Proteins/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Young Adult
7.
J Exp Clin Cancer Res ; 39(1): 23, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31992359

ABSTRACT

BACKGROUND: Tripartite motif-containing proteins (TRIM) play a crucial role in carcinogenesis. Little attention has been focused on the possible functions of TRIM6 on carcinogenesis. METHODS: The expression levels of TRIM6 were assessed in colorectal cancer (CRC) samples. TRIM6 expression was knocked down in CRC cell lines, and subjected to Cell counting kit-8 (CCK-8), bromodeoxyuridine (BrdU) incorporation and cell cycle assays. Immunoprecipitation and proteomics analysis was performed to identify potential associated proteins of TRIM6. RESULTS: TRIM6 expression was up-regulated in CRC samples and TRIM6 expression may be an independent prognostic marker for CRC. Knocking down TRIM6 expression suppressed CRC cell proliferation, induced cell cycle arrested at G2/M phase and increased sensitivity to 5-fluorouracil and oxaliplatin. TIS21, an anti-proliferative protein involved in the regulation of G2/M arrest, was identified as an interaction partner of TRIM6. Moreover, CRC cells with TRIM6 overexpression showed decreased TIS21 protein stability. TIS21 ubiquitination was increased in CRC cells overexpressing TRIM6, but not in those overexpressing TRIM6 E3 catalytic mutant (C15A). Further, Lys5 was essential for TRIM6 mediated TIS21 ubiquitination. TIS21 overexpression reversed the induced effects of TRIM6 overexpression on CRC cell proliferation, and the levels of forkhead box M1 (FoxM1), phosphorylated FoxM1, Cyclin B1 and c-Myc. Thiostrepton, a specific inhibitor for FoxM1, was less effective in anti-proliferative activity against CRC cells with lower level of TRIM6 in vitro and in vivo. CONCLUSIONS: Our study suggests that TRIM6 promotes the progression of CRC via TIS21/FoxM1.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Forkhead Box Protein M1/metabolism , Immediate-Early Proteins/metabolism , Thiostrepton/pharmacology , Tripartite Motif Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Aged , Animals , Anti-Bacterial Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , G2 Phase Cell Cycle Checkpoints/drug effects , HCT116 Cells , HEK293 Cells , Humans , M Phase Cell Cycle Checkpoints/drug effects , Male , Mice , Mice, Nude , Middle Aged , Random Allocation , Tripartite Motif Proteins/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis
8.
Am J Physiol Cell Physiol ; 318(3): C695-C703, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31967859

ABSTRACT

The dysregulation of ubiquitin ligase is the cause of many human diseases. Tripartite motif protein 32 (TRIM32) is an E3 ubiquitin ligase whose role in nucleus pulposus (NP) cell apoptosis is unclear. The expression of TRIM family protein and ß-catenin in 40 NP tissue samples was detected by RT-PCR. Interleukin (IL)-1ß or tumor necrosis factor (TNF)-α was used to treat rat NP cells. Knockdown and overexpression of Trim32 were achieved using specific siRNA and recombinant plasmids. Western blotting, RT-PCR, and flow cytometry were used to assess the expression of TRIM32/ß-catenin and the apoptosis rate of NP cells. Coimmunoprecipitation was adopted to analyze the possible interactions between AXIN1 and TRIM32. In clinical samples, TRIM32 expression was of positive relevance with the expression of CTNNB1 (ß-catenin). In vitro, apoptosis of IL-1ß- or TNF-α-treated rat NP cells was induced through upregulated Trim32 expression and activated ß-catenin signaling, whereas Trim32 siRNA and inhibition of ß-catenin reversed the induction effect of cytokines. Further studies indicated that TRIM32 activated the ß-catenin signaling pathway through ubiquitination of AXIN1, thereby regulating apoptosis. Collectively, this study reveals that TRIM32 promotes inflammatory factor-induced apoptosis of rat NP cells, in part by direct degradation of AXIN1 to trigger ß-catenin signaling.


Subject(s)
Apoptosis/physiology , Axin Protein/metabolism , Nucleus Pulposus/metabolism , Transcription Factors/biosynthesis , Tripartite Motif Proteins/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , beta Catenin/metabolism , Adult , Animals , Cells, Cultured , Female , Humans , Male , Middle Aged , Nucleus Pulposus/pathology , Rats , Rats, Sprague-Dawley , Ubiquitination/physiology
9.
Cancer Biother Radiopharm ; 35(1): 41-49, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31916845

ABSTRACT

Background: NRAGE (neurotrophin receptor-interacting melanoma antigen-encoding gene homolog) has a complex role and regulates cell growth in different tumor cells. Although NRAGE was been discovered for more than 10 years ago, the function of NRAGE in hepatoblastoma (HB) cells is currently unknown. Materials and Methods: The expression of NRAGE was detected by reverse transcription-quantitative polymerase chain reaction assay or western blotting assay. Cellular apoptosis was analyzed to estimate the effect of NRAGE under radiation. The ability of clonogenic capacity was evaluated to confirm the influence of proliferation for NRAGE by radiation. The immunofluorescence assay was used to further study the expression of NRAGE under radiation. A nude mouse tumor xenograft model was constructed to confirm the effect of NRAGE deficiency under radiation conditions in vivo. Results: The authors determined that deletion of NRAGE significantly inhibited HB cell proliferation in vitro and in vivo, and NRAGE knockdown apparently sensitized HB cells to ionizing radiation (IR). Further mechanistic studies revealed that NRAGE plays a critical role in homologous recombination by inhibiting the expression of RNF8 (ring finger protein 8) and BARD1 (BRCA1 associated RING domain 1) and the recruitment of RAD51. Conclusions: The authors demonstrated that downregulation of NRAGE sensitizes HB cell lines to IR in vitro and in vivo. It provides a promising therapeutic strategy for HB patients by specifically targeting NRAGE.


Subject(s)
Antigens, Neoplasm/biosynthesis , Hepatoblastoma/genetics , Hepatoblastoma/radiotherapy , Liver Neoplasms/genetics , Liver Neoplasms/radiotherapy , Neoplasm Proteins/biosynthesis , Recombinational DNA Repair , Animals , Antigens, Neoplasm/genetics , Apoptosis/radiation effects , Cell Line, Tumor , Cell Proliferation/radiation effects , Down-Regulation , Gene Knockdown Techniques , HEK293 Cells , Hep G2 Cells , Hepatoblastoma/metabolism , Hepatoblastoma/pathology , Heterografts , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , Neoplasm Proteins/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transfection , Tripartite Motif Proteins/antagonists & inhibitors , Tripartite Motif Proteins/biosynthesis , Tripartite Motif Proteins/genetics , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
10.
Sci Rep ; 9(1): 19619, 2019 12 23.
Article in English | MEDLINE | ID: mdl-31873114

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths worldwide, with 50-70% of patients suffering from bone metastasis. Accumulating evidence has demonstrated that miRNAs are involved in cell proliferation, migration, and invasion in malignancy, such as lung cancer bone metastasis. In the present study, we demonstrated that reduced miR-192-5p and increased TRIM44 levels were associated with the proliferation, migration and invasion of lung cancer. Furthermore, the potential functions of miR-192-5p were explored in A549 and NCI-H1299 cells. We found that miR-192-5p upregulation suppressed tumour behaviours in lung cancer cells. To further investigate whether miR-192-5p is associated with TRIM44, we used TargetScan software to predict the binding site between miR-192-5p and TRIM44. Luciferase activity assays were performed to verify this prediction. In addition, the significant role of miR-192-5p in negatively regulating TRIM44 expression was manifested by our research group. our results suggest that miR-192-5p inhibited the proliferation, migration and invasion of lung cancer through TRIM44.


Subject(s)
Bone Neoplasms/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Lung Neoplasms/metabolism , MicroRNAs/metabolism , Neoplasm Proteins/biosynthesis , RNA, Neoplasm/metabolism , Tripartite Motif Proteins/biosynthesis , A549 Cells , Animals , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/secondary , Cell Movement , Female , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/genetics , RNA, Neoplasm/genetics , Tripartite Motif Proteins/genetics
11.
Exp Physiol ; 104(10): 1505-1517, 2019 10.
Article in English | MEDLINE | ID: mdl-31357248

ABSTRACT

NEW FINDINGS: What is the central question of this study? What are the adaptations of protein synthesis and degradation that occur in skeletal muscle in response to high-intensity interval training (HIIT), and what are the magnitudes of the changes in response to HIIT, compared to moderate-intensity continuous training (MICT), and the mechanisms underlying these changes? What is the main finding and its importance? HIIT is more effective than MICT in altering the expression of muscle RING-finger protein-1 and muscle atrophy F-box, and enhancing the autophagic flux in rat soleus muscle. In addition, HIIT could activate the mechanistic target of rapamycin pathway. These findings suggest that HIIT might be an effective exercise strategy for health promotion in skeletal muscle. ABSTRACT: This study aimed to investigate the impact of high-intensity interval training (HIIT) on the proteins involved in protein synthesis, the ubiquitin-proteasome system (UPS) and autophagy in skeletal muscle of middle-aged rats. Nine-month-old male Wistar rats (n = 56) were randomly divided into three groups: a control (C) group, a moderate-intensity continuous training (MICT) group and a HIIT group. Rats in the training groups ran on treadmills 5 days per week for 8 weeks. The MICT group ran for 50 min at 60% V̇O2max , while the HIIT group ran for 3 min at 80% of V̇O2max six times separated by 3-min periods at 40% V̇O2max . Aerobic endurance, number of autophagosomes and expression of proteins involved in protein synthesis and degradation in the soleus muscle were measured at three time points: before training, after 4 weeks and after 8 weeks of training. Compared to the C group, HIIT and MICT increased the expression of phosphorylated mechanistic target of rapamycin (mTOR) after 8 weeks (P < 0.05 and P < 0.01, respectively). HIIT increased the expression of muscle RING-finger protein-1 (MuRF-1) after 4 weeks (P < 0.01), and decreased its expression after 8 weeks (P < 0.01). Both HIIT and MICT decreased the expression of muscle atrophy F-box (MAFbx) after 4 weeks (P < 0.05). HIIT improved the expression of microtubule-associated protein 1A/1B-light chain 3 (LC3)-II (P < 0.05), and decreased the P62 content (P < 0.01) after 4 weeks. The LC3II/LC3I ratio was increased after 8 weeks (P < 0.01). This study demonstrated that HIIT could activate the mTOR pathway, alter the expression of MuRF-1 and MAFbx proteins, and enhance autophagic flux in soleus muscle of middle-aged rats.


Subject(s)
Autophagy/physiology , High-Intensity Interval Training , Muscle Proteins/biosynthesis , Muscle, Skeletal/physiology , SKP Cullin F-Box Protein Ligases/biosynthesis , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism , Tripartite Motif Proteins/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Anaerobic Threshold , Animals , Lysosomes/metabolism , Male , Phagosomes/metabolism , Phosphorylation , Rats , Rats, Wistar , Ubiquitin-Specific Proteases/metabolism
12.
FEBS Open Bio ; 9(8): 1413-1420, 2019 08.
Article in English | MEDLINE | ID: mdl-31150153

ABSTRACT

Tripartite motif-containing 14 (TRIM14) is a mitochondrial adaptor that promotes innate immune signaling and plays important roles in antiviral defense. Expression of TRIM14 is induced by interferon (IFN)-I. However, the mechanism by which IFN-I induces TRIM14 production is not yet determined. In this study, we have examined the function of TRIM14 promoter and found that a GC box and an IFN-stimulated response element (ISRE) are necessary for the basal level transcription of TRIM14. We further observed that IFN-I activates the TRIM14 promoter through the ISRE. In particular, interferon regulatory factor (IRF)-1 and IRF-2 bind to the TRIM14 promoter and activate transcription of TRIM14. Moreover, knockdown of IRF-1 reduces the stimulation of TRIM14 transcription by IFN-α, suggesting that IRF-1 is involved in the activation of TRIM14 by IFN-I. IRF-2 has little effect on IFN-α-induced TRIM14 transcription but is essential for the basal transcription of TRIM14.


Subject(s)
Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-2/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , Tripartite Motif Proteins/biosynthesis , Gene Expression Regulation/genetics , HeLa Cells , Humans , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-2/genetics , Interferon-alpha/genetics , Interferon-alpha/metabolism , Interferons/genetics , Interferons/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Promoter Regions, Genetic/genetics , Response Elements/genetics , Signal Transduction , Transcription, Genetic/genetics , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism
13.
Cell Rep ; 27(2): 549-560.e6, 2019 04 09.
Article in English | MEDLINE | ID: mdl-30970257

ABSTRACT

Excessive and unresolved neuroinflammation is a key component of the pathological cascade in brain injuries such as ischemic stroke. Here, we report that TRIM9, a brain-specific tripartite motif (TRIM) protein, was highly expressed in the peri-infarct areas shortly after ischemic insults in mice, but expression was decreased in aged mice, which are known to have increased neuroinflammation after stroke. Mechanistically, TRIM9 sequestered ß-transducin repeat-containing protein (ß-TrCP) from the Skp-Cullin-F-box ubiquitin ligase complex, blocking IκBα degradation and thereby dampening nuclear factor κB (NF-κB)-dependent proinflammatory mediator production and immune cell infiltration to limit neuroinflammation. Consequently, Trim9-deficient mice were highly vulnerable to ischemia, manifesting uncontrolled neuroinflammation and exacerbated neuropathological outcomes. Systemic administration of a recombinant TRIM9 adeno-associated virus that drove brain-wide TRIM9 expression effectively resolved neuroinflammation and alleviated neuronal death, especially in aged mice. These findings reveal that TRIM9 is essential for resolving NF-κB-dependent neuroinflammation to promote recovery and repair after brain injury and may represent an attractive therapeutic target.


Subject(s)
Brain Ischemia/metabolism , Nerve Tissue Proteins/biosynthesis , Stroke/metabolism , Tripartite Motif Proteins/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Animals , Brain Ischemia/genetics , Brain Ischemia/pathology , Cells, Cultured , Female , HEK293 Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Neurons/metabolism , Neurons/pathology , Neuroprotection , Phosphorylation , Stroke/genetics , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
14.
J Am Heart Assoc ; 8(4): e009960, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30741589

ABSTRACT

Background The aortic valve of the heart experiences constant mechanical stress under physiological conditions. Maladaptive valve injury responses contribute to the development of valvular heart disease. Here, we test the hypothesis that MG 53 (mitsugumin 53), an essential cell membrane repair protein, can protect valvular cells from injury and fibrocalcific remodeling processes associated with valvular heart disease. Methods and Results We found that MG 53 is expressed in pig and human patient aortic valves and observed aortic valve disease in aged Mg53-/- mice. Aortic valves of Mg53-/- mice showed compromised cell membrane integrity. In vitro studies demonstrated that recombinant human MG 53 protein protects primary valve interstitial cells from mechanical injury and that, in addition to mediating membrane repair, recombinant human MG 53 can enter valve interstitial cells and suppress transforming growth factor-ß-dependent activation of fibrocalcific signaling. Conclusions Together, our data characterize valve interstitial cell membrane repair as a novel mechanism of protection against valvular remodeling and assess potential in vivo roles of MG 53 in preventing valvular heart disease.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/metabolism , Calcinosis/metabolism , Tripartite Motif Proteins/biosynthesis , Ventricular Remodeling , Animals , Aortic Valve/pathology , Aortic Valve Stenosis/diagnosis , Aortic Valve Stenosis/physiopathology , Biomarkers/metabolism , Blotting, Western , Calcinosis/diagnosis , Calcinosis/physiopathology , Cells, Cultured , Disease Models, Animal , Echocardiography , Humans , Immunohistochemistry , Male , Mice , Signal Transduction , Stress, Mechanical , Swine
15.
Med Sci Monit ; 25: 771-777, 2019 Jan 27.
Article in English | MEDLINE | ID: mdl-30685767

ABSTRACT

BACKGROUND Tripartite motif containing 55 (TRIM55) plays a regulatory role in assembly of sarcomeres, but few studies have assessed its function in hepatocellular carcinoma (HCC). MATERIAL AND METHODS Immunohistochemistry (IHC) was used to detect expression of TRIM55 in tissues samples of HCC patients. Transwell assay was used to study migration and invasion ability of HCC cells. Western blot and immunofluorescence (IF) were used to analyze mechanism of TRIM55 in cell migration and invasion. RESULTS We found TRIM55 was downregulated in HCC tissues and was associated with prognosis of HCC patients. Cox regression analysis showed that TRIM55 was an independent risk factor of prognosis of HCC patients. Overexpression of TRIM55 was associated with lower cell migration and invasion ability, and it led to high expression of E-cadherin and low expression of Vimentin and MMP2. CONCLUSIONS Our study found TRIM55 is an independent factor affecting the prognosis of HCC patients, and overexpression of TRIM55 inhibits migration and invasion of HCC cells through epithelial-mesenchymal transition and MMP2.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Matrix Metalloproteinase 2/metabolism , Tripartite Motif Proteins/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Down-Regulation , Epithelial-Mesenchymal Transition , Female , Humans , Kaplan-Meier Estimate , Liver Neoplasms/genetics , Male , Middle Aged , Neoplasm Invasiveness , Prognosis , Proportional Hazards Models , Signal Transduction , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
16.
Proc Natl Acad Sci U S A ; 115(28): E6566-E6575, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29946036

ABSTRACT

Myosin is a molecular motor indispensable for body movement and heart contractility. Apart from pure cardiomyopathy, mutations in MYH7 encoding slow/ß-cardiac myosin heavy chain also cause skeletal muscle disease with or without cardiac involvement. Mutations within the α-helical rod domain of MYH7 are mainly associated with Laing distal myopathy. To investigate the mechanisms underlying the pathology of the recurrent causative MYH7 mutation (K1729del), we have developed a Drosophila melanogaster model of Laing distal myopathy by genomic engineering of the Drosophila Mhc locus. Homozygous MhcK1728del animals die during larval/pupal stages, and both homozygous and heterozygous larvae display reduced muscle function. Flies expressing only MhcK1728del in indirect flight and jump muscles, and heterozygous MhcK1728del animals, were flightless, with reduced movement and decreased lifespan. Sarcomeres of MhcK1728del mutant indirect flight muscles and larval body wall muscles were disrupted with clearly disorganized muscle filaments. Homozygous MhcK1728del larvae also demonstrated structural and functional impairments in heart muscle, which were not observed in heterozygous animals, indicating a dose-dependent effect of the mutated allele. The impaired jump and flight ability and the myopathy of indirect flight and leg muscles associated with MhcK1728del were fully suppressed by expression of Abba/Thin, an E3-ligase that is essential for maintaining sarcomere integrity. This model of Laing distal myopathy in Drosophila recapitulates certain morphological phenotypic features seen in Laing distal myopathy patients with the recurrent K1729del mutation. Our observations that Abba/Thin modulates these phenotypes suggest that manipulation of Abba/Thin activity levels may be beneficial in Laing distal myopathy.


Subject(s)
Distal Myopathies , Drosophila Proteins/metabolism , Genetic Loci , Mutation , Myocardium/metabolism , Myosin Heavy Chains , Tripartite Motif Proteins , Animals , Disease Models, Animal , Distal Myopathies/genetics , Distal Myopathies/metabolism , Distal Myopathies/pathology , Drosophila Proteins/genetics , Drosophila melanogaster , Homozygote , Humans , Myocardium/pathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Tripartite Motif Proteins/biosynthesis , Tripartite Motif Proteins/genetics
17.
Muscle Nerve ; 58(2): 314-318, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29572868

ABSTRACT

INTRODUCTION: Apigenin (AP) has been reported to elicit anti-inflammatory effects. In this study, we investigated the effect of AP on sciatic nerve denervation-induced muscle atrophy. METHODS: Sciatic nerve-denervated mice were fed a 0.1% AP-containing diet for 2 weeks. Muscle weight and cross-sectional area (CSA), and the expression of atrophic genes and inflammatory cytokines in the gastrocnemius were analyzed. RESULTS: Denervation significantly induced muscle atrophy. However, values for muscle weight and CSA were greater in the denervated muscle of the AP mice than the controls. AP suppressed the expression of MuRF1, but upregulated both myosin heavy chain (MHC) and MHC type IIb. AP also significantly suppressed expression of tumor necrosis-alpha in the gastrocnemius and soleus muscles, and interleukin-6 expression in the soleus muscle. DISCUSSION: AP appears to inhibit denervation-induced muscle atrophy, which may be due in part to its inhibitory effect on inflammatory processes within muscle. Muscle Nerve 58: 314-318, 2018.


Subject(s)
Apigenin/therapeutic use , Muscular Atrophy/etiology , Muscular Atrophy/prevention & control , Sciatic Nerve , Anatomy, Cross-Sectional , Animals , Denervation , Gene Expression/drug effects , Interleukin-6/biosynthesis , Interleukin-6/genetics , Male , Mice , Mice, Inbred C57BL , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Muscular Atrophy/genetics , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/genetics , Organ Size , Tripartite Motif Proteins/biosynthesis , Tripartite Motif Proteins/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/genetics
18.
Gut ; 67(1): 166-178, 2018 01.
Article in English | MEDLINE | ID: mdl-28341749

ABSTRACT

OBJECTIVE: Interferons (IFNs) mediate direct antiviral activity. They play a crucial role in the early host immune response against viral infections. However, IFN therapy for HBV infection is less effective than for other viral infections. DESIGN: We explored the cellular targets of HBV in response to IFNs using proteome-wide screening. RESULTS: Using LC-MS/MS, we identified proteins downregulated and upregulated by IFN treatment in HBV X protein (HBx)-stable and control cells. We found several IFN-stimulated genes downregulated by HBx, including TRIM22, which is known as an antiretroviral protein. We demonstrated that HBx suppresses the transcription of TRIM22 through a single CpG methylation in its 5'-UTR, which further reduces the IFN regulatory factor-1 binding affinity, thereby suppressing the IFN-stimulated induction of TRIM22. CONCLUSIONS: We verified our findings using a mouse model, primary human hepatocytes and human liver tissues. Our data elucidate a mechanism by which HBV evades the host innate immune system.


Subject(s)
5' Untranslated Regions/genetics , CpG Islands/genetics , Hepatitis B virus/immunology , Interferons/immunology , Minor Histocompatibility Antigens/genetics , Repressor Proteins/genetics , Tripartite Motif Proteins/genetics , Animals , Down-Regulation/genetics , Down-Regulation/immunology , Epigenesis, Genetic , Gene Expression Regulation/immunology , Hepatocytes/metabolism , Humans , Immune Evasion , Liver/metabolism , Methylation , Mice , Minor Histocompatibility Antigens/biosynthesis , Proteome , Repressor Proteins/biosynthesis , Tripartite Motif Proteins/biosynthesis
19.
Mol Cell Biochem ; 444(1-2): 161-168, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29198019

ABSTRACT

Skeletal muscle atrophy occurs in different catabolic conditions and mostly accompanied with upregulation of Muscle ring finger 1 (MuRF1) gene which is one of the master regulatory genes in muscle atrophy. Taurine amino acid is widely distributed in different tissues and has anti-inflammatory and antioxidant effects. This study aimed to investigate the potential influence of taurine on muscle atrophy induced by reduced mechanical loading. Twenty-eight Albino mice were used, and divided equally into four groups: group I (control); group II (immobilization); group III (immobilization + taurine); and group IV (taurine). Quadriceps muscle sections were taken for histopathology, immunohistochemical analysis of caspase 3 expression, and qRT-PCR of MuRF1 gene. Our data revealed Zenker necrosis associated with axonal injury of the nerve trunk of the immobilized muscle together with increase of caspase 3 expression and upregulation of MuRF1 gene. While, taurine supplementation alleviated the muscular and neural tissues damage associated with disuse skeletal muscle atrophy through downregulation of MuRF1 gene and decrease of tissue caspase 3 expression. In conclusion, taurine may be helpful to counteract apoptosis and up-regulated MuRF1 gene expression related to muscle atrophy, which might be hopeful for a large number of patients.


Subject(s)
Muscle Proteins/biosynthesis , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Proteolysis/drug effects , Taurine/pharmacology , Tripartite Motif Proteins/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Up-Regulation/drug effects , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Male , Mice , Muscle, Skeletal/pathology , Muscular Atrophy/pathology
20.
Biosci Rep ; 37(4)2017 Aug 31.
Article in English | MEDLINE | ID: mdl-28620119

ABSTRACT

Tripartite motif containing 25 (TRIM25) is a member of TRIM proteins and functions as an E3 (ubiquitin ligase). It has been found to act as an oncogene in gastric cancer cells and is abnormally expressed in cancers in female reproductive system. Here, we investigated the function of TRIM25 in colorectal cancer. TRIM25 was found to be significantly up-regulated in colorectal cancer tissues and cancer cell lines through real-time PCR assay. Colorectal cancer cells (CRCs) overexpressing TRIM25 exhibited a two-fold higher proliferation and migration rate compared with their parental lines in vitro Moreover, TRIM25 also promoted tumor progression in vivo Further study indicated that TRIM25 worked through positively regulating transforming growth factor ß (TGF-ß) signaling pathway to regulate the proliferation and invasion of CRCs. In summary, our results indicate that TRIM25 also acts as an oncogene in colorectal cancer and it functions through TGF-ß signaling pathway. Thus, TRIM25 represents potential targets for the treatment of colorectal cancer.


Subject(s)
Cell Proliferation , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Signal Transduction , Transcription Factors/biosynthesis , Tripartite Motif Proteins/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Up-Regulation , Cell Line, Tumor , Colorectal Neoplasms/pathology , Female , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...