Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
J Biol Chem ; 298(10): 102339, 2022 10.
Article in English | MEDLINE | ID: mdl-35931121

ABSTRACT

Family with sequence similarity 83 A (FAM83A) is a newly discovered proto-oncogene that has been shown to play key roles in various cancers. However, the function of FAM83A in other physiological processes is not well known. Here, we report a novel function of FAM83A in adipocyte differentiation. We used an adipocyte-targeting fusion oligopeptide (FITC-ATS-9R) to deliver a FAM83A-sgRNA/Cas9 plasmid to knockdown Fam83a (ATS/sg-FAM83A) in white adipose tissue in mice, which resulted in reduced white adipose tissue mass, smaller adipocytes, and mitochondrial damage that was aggravated by a high-fat diet. In cultured 3T3-L1 adipocytes, we found loss or knockdown of Fam83a significantly repressed lipid droplet formation and downregulated the expression of lipogenic genes and proteins. Furthermore, inhibition of Fam83a decreased mitochondrial ATP production through blockage of the electron transport chain, associated with enhanced apoptosis. Mechanistically, we demonstrate FAM83A interacts with casein kinase 1 (CK1) and promotes the permeability of the mitochondrial outer membrane. Furthermore, loss of Fam83a in adipocytes hampered the formation of the TOM40 complex and impeded CK1-driven lipogenesis. Taken together, these results establish FAM83A as a critical regulator of mitochondria maintenance during adipogenesis.


Subject(s)
Adipocytes, White , Adipogenesis , Casein Kinase I , Mitochondria , Neoplasm Proteins , Proto-Oncogenes , Animals , Mice , 3T3-L1 Cells , Adipocytes, White/cytology , Adipocytes, White/metabolism , Adipogenesis/genetics , Casein Kinase I/metabolism , Cell Differentiation , Mitochondria/genetics , Mitochondria/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism
2.
J Biol Chem ; 296: 100037, 2021.
Article in English | MEDLINE | ID: mdl-33158991

ABSTRACT

With the improvement of people's living standards, the number of obese patients has also grown rapidly. It is reported that the level of oxidative stress in obese patients has significantly increased, mainly caused by the increase in reactive oxygen species (ROS) levels in adipose tissue. Studies have shown that the use of siRNA to interfere with bone morphogenetic protein and activin membrane-bound inhibitor (BAMBI) expression could promote adipocyte differentiation, and under hypoxic conditions, BAMBI could act as a regulator of HIF1α to regulate the polarity damage of epithelial cells. In view of these results, we speculated that BAMBI may regulate adipogenesis by regulating the level of ROS. In this study, we generated adipose-specific BAMBI knockout mice (BAMBI AKO) and found that compared with control mice, BAMBI AKO mice showed obesity when fed with high-fat diet, accompanied by insulin resistance, glucose intolerance, hypercholesterolemia, and increased inflammation in adipose tissue. Interestingly, adipose-specific deficiency of BAMBI could cause an increase in the expression level of Nox4, thereby promoting ROS production in cytoplasm and mitochondria and the DNA-binding activity of C/EBPß and ultimately promoting adipogenesis. Consistently, our findings indicated that BAMBI may be a reactive oxygen regulator to affect adipogenesis, thereby controlling obesity and metabolic syndrome.


Subject(s)
Adipogenesis , Adipose Tissue/metabolism , Bone Morphogenetic Proteins/metabolism , Membrane Proteins/genetics , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Adipose Tissue/cytology , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Diet, High-Fat , Fatty Liver/genetics , Humans , Insulin Resistance/genetics , Mice , Mice, Knockout
3.
J Fluoresc ; 31(2): 385-392, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33403518

ABSTRACT

A new three-dimensional lanthanide metal-organic framework (Ln-MOF), [Eu4(L)4(H2O)8]·10H2O (1, H3L = biphenyl-3'-nitro-3,4',5-tricarboxylic acid), has been constructed via solvothermal technology and its framework has been detected by the single-crystal X-ray diffraction and elemental analyses. Complex 1 with typical emission of Eu3+ ion represents dramatic luminescence quenching affect for picric acid (PA) and the linear Stern-Volmer plot was surveyed in the consistence, ranging from 0.05 to 0.15 mM (Ksv = 98,074 M- 1). Its therapeutic effect of the compound on the cerebral edema caused by cerebral hemorrhage was estimated and the mechanism was explored. Possible binding interactions have been investigated by molecular docking simulations, from which the binding interactions are identified and the carboxyl oxygens are responsible for those identified interactions.


Subject(s)
Brain Edema/diagnosis , Cerebral Hemorrhage/diagnosis , Coordination Complexes/chemistry , Europium/chemistry , Metal-Organic Frameworks/chemistry , Picrates/analysis , Animals , Brain Edema/metabolism , Cerebral Hemorrhage/metabolism , Coordination Complexes/chemical synthesis , Enzyme-Linked Immunosorbent Assay , Metal-Organic Frameworks/chemical synthesis , Models, Molecular , Oxidation-Reduction , Prothrombin/metabolism , Rats , Rats, Sprague-Dawley
4.
J Anim Physiol Anim Nutr (Berl) ; 104(3): 954-964, 2020 May.
Article in English | MEDLINE | ID: mdl-32056287

ABSTRACT

MicroRNA (miRNA) is essential for the process of gene posttranscriptional regulation in skeletal muscle of many species, such as mice, cattle and so on. However, a little number of miRNAs have been reported in the muscle development of Chinese native pig breeds. In this study, the longissimus dorsi transcripts of Chinese native Rongchang pig at weaning and slaughter time points were analysed for miRNA-seq. The results showed that 19 novel and 186 known miRNAs involved in the Rongchang pig skeletal muscle development were identified. Based on these findings, we further confirmed that porcine miR-127, miR-299 and miR-432-5p were obviously down-expressed in adult pig (287 days of age), while miR-7134-3p and 664-5p were significantly up-expressed in weaning pig (35 days of age). In other words, these miRNAs could be the potential molecular markers and play vital roles in the muscle development process. Moreover, we found miR-127 could inhibit the proliferation and myogenesis of porcine satellite cells in longissimus dorsi muscle. Our findings will provide deep insight into miRNA function for pork quality research with Chinese indigenous pig breeds.


Subject(s)
Aging/physiology , MicroRNAs/metabolism , Muscle, Skeletal/metabolism , Swine/physiology , Animals , Animals, Newborn , Cell Proliferation , Cells, Cultured , Gene Expression Regulation , MicroRNAs/genetics , Satellite Cells, Skeletal Muscle/physiology , Transcriptome
5.
J Biol Chem ; 293(50): 19177-19190, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30323063

ABSTRACT

MicroRNAs (miRNAs) are noncoding RNAs that regulate gene expression at the post-transcriptional level and are involved in the regulation of the formation, maintenance, and function of skeletal muscle. Using miRNA sequencing and bioinformatics analysis, we previously found that the miRNA miR-664-5p is significantly differentially expressed in longissimus dorsi muscles of Rongchang pigs. However, the molecular mechanism by which miR-664-5p regulates myogenesis remains unclear. In this study, using flow cytometry, 5-ethynyl-2'-deoxyuridine staining, and cell count and immunofluorescent assays, we found that cell-transfected miR-664-5p mimics greatly promoted proliferation of C2C12 mouse myoblasts by increasing the proportion of cells in the S- and G2-phases and up-regulating the expression of cell cycle genes. Moreover, miR-664-5p inhibited myoblast differentiation by down-regulating myogenic gene expression. In contrast, miR-664-5p inhibitor repressed myoblast proliferation and promoted myoblast differentiation. Mechanistically, using dual-luciferase reporter gene experiments, we demonstrated that miR-664-5p directly targets the 3'-UTR of serum response factor (SRF) and Wnt1 mRNAs. We also observed that miR-664-5p inhibits both mRNA and protein levels of SRF and Wnt1 during myoblast proliferation and myogenic differentiation, respectively. Furthermore, the activating effect of miR-664-5p on myoblast proliferation was attenuated by SRF overexpression, and miR-664-5p repressed myogenic differentiation by diminishing the accumulation of nuclear ß-catenin. Of note, miR-664-5p's inhibitory effect on myogenic differentiation was abrogated by treatment with Wnt1 protein, the key activator of the Wnt/ß-catenin signaling pathway. Collectively, our findings suggest that miR-664-5p controls SRF and canonical Wnt/ß-catenin signaling pathways in myogenesis.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation/genetics , MicroRNAs/metabolism , Myoblasts/metabolism , Serum Response Factor/metabolism , Wnt1 Protein/metabolism , Animals , Down-Regulation , HEK293 Cells , Humans , Mice , Muscle Development/genetics , RNA, Messenger/genetics , Serum Response Factor/genetics , Wnt Signaling Pathway , Wnt1 Protein/genetics
6.
J Cell Physiol ; 234(12): 21838-21850, 2019 12.
Article in English | MEDLINE | ID: mdl-31206669

ABSTRACT

MicroRNAs (miRNAs) have critical roles during adipogenesis; however, their precise functions are not completely understood. Porcine miRNA expression profiles show that miR-127 is dramatically downregulated with age in adipose tissue. We aimed to identify the precise functions and mechanisms of miR-127 in proliferation and adipogenesis. Preadipocytes were cultured under conditions to induce proliferation or differentiation and the effect of miR-127 overexpression on these processes, and the associated bioinformatically predicted target genes, were assessed using luciferase assays, quantitative real-time PCR, western blot analysis, and cell staining techniques. miR-127 increased proliferation by promoting cell cycling, whereas it suppressed differentiation, which was accompanied by reduced lipid accumulation. miR-127 targeted mitogen-activated protein kinase 4 and homeobox C6 (HOXC6) to activate preadipocyte proliferation. During differentiation, miR-127 targeted HOXC6 to attenuate adipogenesis. These findings identify miR-127 as an inhibitor of porcine adipogenesis, which may inform future strategies to reduce porcine fat deposition and treat human obesity.


Subject(s)
Adipocytes/metabolism , Adipogenesis/genetics , Cell Differentiation/genetics , Cell Proliferation/genetics , Genes, Homeobox/genetics , Animals , Homeodomain Proteins/genetics , Mitogen-Activated Protein Kinases/metabolism , Swine
7.
RNA Biol ; 14(3): 347-360, 2017 03 04.
Article in English | MEDLINE | ID: mdl-28085550

ABSTRACT

Skeletal muscle is the dominant executant in locomotion and regulator in energy metabolism. Embryonic myogenesis and postnatal muscle growth are controlled by a cascade of transcription factors and epigenetic regulatory mechanisms. MicroRNAs (miRNAs), a family of non-coding RNA of 22 nucleotides in length, post-transcriptionally regulates expression of mRNA by pairing the seed sequence to 3' UTR of target mRNA. Increasing evidence has demonstrated that miRNAs are important regulators in diverse myogenic processes. The profiling of miRNA expression revealed that miR-432 is more enriched in the longissimus dorsi of 35-day-old piglets than that of adult pigs. Our gain of function study showed that miR-432 can negatively regulate both myoblast proliferation and differentiation. Mechanically, we found that miR-432 is able to down-regulate E2F transcription factor 3 (E2F3) to inactivate the expression of cell cycle and myogenic genes. We also identified that phosphatidylinositol 3-kinase regulatory subunit (P55PIK) is another target gene of miR-432 in muscle cells. downregulation of P55PIK by miR-432 leads to inhibition of P55PIK-mediated PI3K/AKT/mTOR signaling pathway during differentiation. The blocking effect of miR-432 on this pathway can be rescued by insulin treatment. Taken together, our findings identified microRNA-432 as a potent inhibitor of myogenesis which functions by targeting E2F3 and P55PIK in muscle cells.


Subject(s)
E2F3 Transcription Factor/genetics , MicroRNAs/genetics , Muscle Development/physiology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , 3' Untranslated Regions , Animals , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , Cluster Analysis , E2F3 Transcription Factor/metabolism , Gene Expression Profiling , Gene Expression Regulation , Male , Mice , Models, Biological , Myoblasts/cytology , Myoblasts/metabolism , RNA Interference , Swine , Transcriptome
8.
Exp Cell Res ; 344(1): 11-21, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26940012

ABSTRACT

Intramuscular fat (IMF) has been demonstrated as one of the crucial factors of livestock meat quality. The MAT2B protein with MAT2α catalyzes the formation of methyl donor S- adenosylmethionine (SAMe) to mediate cell metabolism including proliferation and apoptosis. However, the regulatory effect of MAT2B on IMF deposition is still unclear. In this study, the effect of MAT2B on adipogenesis and its potential mechanism during porcine intramuscular preadipocyte differentiation was studied. The results showed that overexpression of MAT2B promoted adipogenesis and significantly up-regulated the mRNA and protein levels of adipogenic marker genes including FASN, PPARγ and aP2, consistently, knockdown of MAT2B inhibited lipid accumulation and down-regulated the mRNA and protein levels of the above genes. Furthermore, flow cytometry and EdU-labeling assay indicated that MAT2B regulate adipogenesis was partly due to influence intracellular SAMe levels and further affect cell clonal expansion. Also, increased expression of MAT2B activated the phosphorylations of AKT and ERK1/2, whereas knockdown of MAT2B blocked AKT signaling and repressed the phosphorylation of ERK1/2. Moreover, the inhibitory effect of LY294002 (a specific PI3K inhibitor) on the activities of AKT and ERK1/2 was partially recovered by overexpression of MAT2B in porcine intramuscular adipocytes. Finally, Co-IP experiments showed that MAT2B can directly interact with AKT. Taken together, our findings suggested that MAT2B acted as a positive regulator through modifying SAMe levels as well as activating AKT/ERK signaling pathway to promote porcine intramuscular adipocyte differentiation.


Subject(s)
Adipocytes/cytology , Adipogenesis , Cell Differentiation , MAP Kinase Signaling System , Methionine Adenosyltransferase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , S-Adenosylmethionine/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Adipogenesis/genetics , Animals , Cell Differentiation/drug effects , Chromones/pharmacology , Gene Expression Profiling , Gene Knockdown Techniques , Intracellular Space/drug effects , Intracellular Space/metabolism , Lentivirus/metabolism , MAP Kinase Signaling System/drug effects , Methionine Adenosyltransferase/genetics , Morpholines/pharmacology , Muscle, Skeletal/cytology , Phosphorylation/drug effects , Protein Binding/drug effects , Sus scrofa
9.
Int J Mol Sci ; 18(11)2017 Nov 15.
Article in English | MEDLINE | ID: mdl-29140299

ABSTRACT

Suitable intramuscular fat (IMF) content improves porcine meat quality. The vital genes regulating IMF deposition are necessary for the selection and breeding of an IMF trait. However, the effect and mechanism of PDGFRα on IMF deposition are still unclear. Here, PDGFRα is moderately expressed in porcine longissimus dorsi muscle (LD), whereas it highly expressed in white adipose tissue (WAT). Moreover, PDGFRα-positive cells were located in the gaps of LD fibers which there were IMF adipocytes. Compared with 180-day-old and lean-type pigs, the levels of PDGFRα were much higher in one-day-old and fat-type pigs. Meanwhile the levels of PDGFRα gradually decreased during IMF preadipocyte differentiation. Furthermore, PDGFRα promoted adipogenic differentiation through activating Erk signaling pathway. Based on PDGFRα upstream regulation analysis, we found that the knockdown of FoxO1 repressed lipogenesis by downregulating PDGFRα, and miR-34a inhibited adipogenesis through targeting PDGFRα. Collectively, PDGFRα is a positive regulator of IMF deposition. Therefore, we suggest that PDGFRα is a possible target to improve meat quality.


Subject(s)
Adipocytes/metabolism , Adipogenesis/genetics , Forkhead Transcription Factors/metabolism , MAP Kinase Signaling System , MicroRNAs/metabolism , Muscles/cytology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Adiposity , Animals , Cell Differentiation/genetics , Gene Expression Profiling , Gene Expression Regulation , Gene Knockdown Techniques , Lipogenesis , MicroRNAs/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Sus scrofa , Time Factors
10.
Acta Biochim Biophys Sin (Shanghai) ; 48(6): 509-19, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27125977

ABSTRACT

The induced lipogenesis and its regulation in C2C12 myoblasts remain largely unclear. Here, we found that the cocktail method could significantly induce lipogenesis through regulating lipid metabolic genes and Erk1/2 phosphorylation in myoblasts. Meanwhile, the expression and secretion of CTRP6 were increased during ectopic lipogenesis. Moreover, CTRP6 knockdown down-regulated the levels of lipogenic genes and phosphorylated Erk1/2 (p-Erk1/2) in the early lipogenic stage, whereas up-regulated p-Erk1/2 in the terminal differentiation. Interestingly, the effect of CTRP6 siRNA was attenuated by U0126 (a special p-Erk1/2 inhibitor) in myoblasts. Furthermore, AdipoR1, not AdipoR2, was first identified as a receptor of CTRP6 during the process of mitotic clonal expansion. Collectively, we suggest that CTRP6 mediates the ectopic lipogenesis through AdipoR1/Erk/PPARγ signaling pathway in myoblasts. Our findings will shed light on the novel biological function of CTRP6 during myoblast lipogenesis and provide a hopeful direction of improving meat quality of domestic animal by lipogenic regulation in skeletal muscle myoblasts.


Subject(s)
Adipokines/metabolism , Lipogenesis/physiology , Myoblasts, Skeletal/metabolism , Adipokines/antagonists & inhibitors , Adipokines/genetics , Animals , Cell Differentiation , Cell Line , Gene Knockdown Techniques , Lipogenesis/genetics , MAP Kinase Signaling System , Mice , Models, Biological , Myoblasts, Skeletal/cytology , PPAR gamma/metabolism , RNA, Small Interfering/genetics , Receptors, Adiponectin/antagonists & inhibitors , Receptors, Adiponectin/genetics , Receptors, Adiponectin/metabolism , Signal Transduction
11.
Zhonghua Zhong Liu Za Zhi ; 37(11): 810-5, 2015 Nov.
Article in Zh | MEDLINE | ID: mdl-26887509

ABSTRACT

OBJECTIVE: To explore the effect of colon cancer cell-derived interleukin-1α on the migration and proliferation of human umbilical vein endothelial cells as well as the role of IL-1α and IL-1ra in the angiogenesis process. METHODS: Western blot was used to detect the expression of IL-1α and IL-1R1 protein in the colon cancer cell lines with different liver metastatic potential. We also examined how IL-1α and IL-1ra influence the proliferation and migration of umbilical vascular endothelial cells assessed by PreMix WST-1 assay and migration assay, respectively. Double layer culture technique was used to detect the effect of IL-1α on the proliferation and migration of vascular endothelial cells and the effect of IL-1ra on the vascular endothelial cells. RESULTS: Western blot analysis showed that IL-1α protein was only detected in highly metastatic colon cancer HT-29 and WiDr cells, but not in the lowly metastatic CaCo-2 and CoLo320 cells.Migration assay showed that there were significant differences in the number of penetrated cells between the control (17.9±3.6) and 1 ng/ml rIL-1α group (23.2±4.2), 10 ng/ml rIL-1α group (31.7±4.5), and 100 ng/ml rIL-1α group (38.6±4.9), showing that it was positively correlated with the increasing concentration of rIL-1α (P<0.01 for all). The proliferation assay showed that the absorbance values were 1.37±0.18 in the control group, and 1.79±0.14 in the 1 ng/ml rIL-1α group, 2.14±0.17 in the 10 ng/ml rIL-1α group, and 2.21±0.23 in the 100 ng/ml rIL-1α group, showing a positive correlation with the increasing concentration of rIL-1α(P<0.01 for all). IL-1ra significantly inhibited the proliferation and migration of vascular endothelial cells (P<0.01). The levels of VEGF protein were (1.697±0.072) ng/ml, (3.507±0.064)ng/ml and (4.139±0.039)ng/ml in the control, HUVECs+ IL-1α and HUVECs+ HT-29 co-culture system groups, respectively, showing a significant difference between the control and HUVECs+ 10 pg/ml rIL-1α groups and between the control and HUVECs+ HT-29 groups (P<0.01 for both). CONCLUSIONS: Our findings indicate that colon cancer cell-derived IL-1α plays an important role in the liver metastasis of colon cancer through increased VEGF level of the colon cancer cells and enhanced vascular endothelial cells proliferation, migration and angiogenesis, while IL-1ra can suppress the effect of IL-1α and inhibit the angiogenesis in colon cancer.


Subject(s)
Colonic Neoplasms/blood supply , Colonic Neoplasms/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Interleukin 1 Receptor Antagonist Protein/physiology , Interleukin-1alpha/physiology , Neovascularization, Pathologic/etiology , Blotting, Western , Caco-2 Cells , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Coculture Techniques , Colonic Neoplasms/pathology , Humans , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin-1alpha/metabolism , Liver Neoplasms/secondary
12.
Cell Signal ; 123: 111356, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39173857

ABSTRACT

Diffuse large B-cell lymphoma (DLBCL) is the most common diagnosed subtype of lymphoma with high invasiveness and heterogeneity. Glycolysis is involved in regulating DLBCL progression. We aimed to explore the role of forkhead box protein A1 (FOXA1) in DLBCL and the mechanisms related to sirtuine5 (SIRT5) and glycolysis. FOXA1 expression in DLBCL cells was analyzed. Then, the proliferation and apoptosis of DLBCL cells were detected using Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EDU) staining and flow cytometry analysis following FOXA1 or SIRT5 knockdown. The glycolysis was assessed by measuring extracellular acidification rate (ECAR), glucose consumption and lactate secretion. Immunoblotting was employed to examine the expression of apoptosis- and glycolysis-related proteins. Additionally, luciferase reporter assay and chromatin immunoprecipitation (ChIP) assay were conducted to test the combination of FOXA1 to SIRT5 promotor region. Subsequently, SIRT5 expression was upregulated to conduct rescue assays. Finally, the effects of FOXA1 downregulation on the growth and glycolysis in OCI-ly7 tumor-bearing mice were examined. As a result, FOXA1 was upregulated in DLBCL cells and FOXA1 or SIRT5 knockdown inhibited the proliferation, accelerated the apoptosis and suppressed glycolysis reprograming in DLBCL cells. Importantly, FOXA1 could transcriptionally activate SIRT5 expression in DLBCL cells. Besides, SIRT5 overexpression counteracted the effects of FOXA1 deficiency on the proliferation, apoptosis and glycolysis reprogramming in DLBCL cells. Furthermore, FOXA1 knockdown inhibited the tumor growth, suppressed the glycolysis reprogramming and downregulated SIRT5 expression in vivo. In summary, FOXA1 could transcriptionally activate SIRT5 to reprogram glycolysis, thereby facilitating the malignant progression of DLBCL.


Subject(s)
Glycolysis , Hepatocyte Nuclear Factor 3-alpha , Lymphoma, Large B-Cell, Diffuse , Sirtuins , Transcriptional Activation , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Animals , Sirtuins/metabolism , Sirtuins/genetics , Mice , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Apoptosis , Disease Progression
13.
Front Biosci (Landmark Ed) ; 29(9): 339, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39344333

ABSTRACT

BACKGROUND: C1q/tumor necrosis factor-related protein-9 (CTRP9) is critically involved in the pathophysiology of metabolic and cardiovascular disorders. This investigation aimed to clarify the mechanism underlying the role of CTRP9 in atherosclerosis in apolipoprotein E (ApoE) knockout (KO) mice. METHODS: ApoE KO mice were fed a Western diet and injected with a virus which resulted in CTRP9 overexpression or knockdown for 12 weeks. The plasma lipid levels and atherosclerotic plaque areas were measured after the mice were euthanized. Aortas were isolated, and RNA sequencing was performed to identify the differentially expressed genes and related signaling pathways. Finally, plasma oxidative stress factors were measured to demonstrate the reliability of the RNA sequencing results. RESULTS: The plasma lipid levels in the CTRP9 overexpression group did not significantly differ from those in the green fluorescence protein (GFP) group. Markablely, CTRP9 overexpression inhibited atherosclerotic plaque formation in ApoE KO mice, whereas CTRP9 knockdown promoted plaque formation. RNA sequencing analysis identified 3485 differentially expressed genes that were prominently enriched across 55 signaling pathways. Additionally, plasma oxidative stress factors were significantly reduced after CTRP9 overexpression, whereas these factors were increased after CTRP9 knockdown, which was consistent with the results of the RNA sequencing analysis. CONCLUSIONS: These findings demonstrated that CTRP9 alleviated inflammation and cholesterol metabolism, which reduced oxidative stress in an atherosclerotic animal model. These beneficial effects may mediate the suppression of lesion development in the aorta.


Subject(s)
Apolipoproteins E , Atherosclerosis , Oxidative Stress , Animals , Atherosclerosis/genetics , Atherosclerosis/metabolism , Male , Mice , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Adiponectin/genetics , Adiponectin/metabolism , Adiponectin/blood , Mice, Knockout, ApoE , Mice, Knockout , Signal Transduction , Plaque, Atherosclerotic/genetics , Plaque, Atherosclerotic/metabolism , Aorta/metabolism , Aorta/pathology , Mice, Inbred C57BL , Adipokines/metabolism , Adipokines/genetics , Lipids/blood , Glycoproteins/genetics , Glycoproteins/metabolism
14.
Exp Ther Med ; 28(2): 328, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38979020

ABSTRACT

There has been interest in the connection between cardiovascular diseases and osteoporosis, both of which share hyperlipidemia as a common pathological basis. Osteoporosis is a progressive metabolic bone disease characterized by reduced bone mass, deteriorated bone microstructure, increased bone fragility and heightened risk of bone fractures. Dysfunction of osteoblastic cells, vital for bone formation, is induced by excessive internalization of lipids under hyperlipidemic conditions, forming the crux of hyperlipidemia-associated osteoporosis. Autophagy, a process fundamental to cell self-regulation, serves a critical role in osteoblastic cell function and bone formation. When activated by lipids, lipophagy inhibits osteoblastic cell differentiation in response to elevated lipid concentrations, resulting in reduced bone mass and osteoporosis. However, an in-depth understanding of the precise roles and mechanisms of lipophagy in the regulation of osteoblastic cell function is required. Study of the molecular mechanisms governing osteoblastic cell response to excessive lipids can result in a clearer understanding of osteoporosis; therefore, potential strategies for preventing hyperlipidemia-induced osteoporosis can be developed. The present review discusses recent progress in elucidating the molecular mechanisms of lipophagy in the regulation of osteoblastic cell function, offering insights into hyperlipidemia-induced osteoporosis.

15.
Exp Ther Med ; 27(1): 28, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38125356

ABSTRACT

BMP and activin membrane-bound inhibitor (BAMBI) is a transmembrane glycoprotein, known as a pseudo-receptor for TGFß, as, while its extracellular domain is similar to that of type I TGFß receptors, its intracellular structure is shorter and lacks a serine/threonine phosphokinase signaling motif. BAMBI can regulate numerous biological phenomena, including glucose and lipid metabolism, inflammatory responses, and cell proliferation and differentiation. Furthermore, abnormal expression of BAMBI at the mRNA and protein levels contributes to various human pathologies, including obesity and cancer. In the present review, the structure of BAMBI is briefly introduced and its associated signaling pathways and physiological functions are described. Understanding of BAMBI structure and function may contribute to knowledge regarding the occurrence of diseases, including obesity and diabetes, among others. The present review provides a theoretical foundation for the development of BAMBI as a potential biomarker or therapeutic target.

16.
Med Biol Eng Comput ; 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39322859

ABSTRACT

With the rapid advancement of modern medical technology, microscopy imaging systems have become one of the key technologies in medical image analysis. However, manual use of microscopes presents issues such as operator dependency, inefficiency, and time consumption. To enhance the efficiency and accuracy of medical image capture and reduce the burden of subsequent quantitative analysis, this paper proposes an improved microscope salient object detection algorithm based on U2-Net, incorporating deep learning technology. The improved algorithm first enhances the network's key information extraction capability by incorporating the Convolutional Block Attention Module (CBAM) into U2-Net. It then optimizes network complexity by constructing a Simple Pyramid Pooling Module (SPPM) and uses Ghost convolution to achieve model lightweighting. Additionally, data augmentation is applied to the slides to improve the algorithm's robustness and generalization. The experimental results show that the size of the improved algorithm model is 72.5 MB, which represents a 56.85% reduction compared to the original U2-Net model size of 168.0 MB. Additionally, the model's prediction accuracy has increased from 92.24 to 97.13%, providing an efficient means for subsequent image processing and analysis tasks in microscopy imaging systems.

17.
J Cancer ; 15(8): 2424-2430, 2024.
Article in English | MEDLINE | ID: mdl-38495482

ABSTRACT

Cofilin (CFL1) is one critical member of the actin deploy family (ADF). Overexpression of CFL1 is associated with aggressive features and poor prognosis in malignancies. We evaluated the expression of CFL1 in patients with chronic myeloid leukemia in the chronic phase (CML-CP), acute myelocytic leukemia (AML) and healthy controls. The role of CFL1 in imatinib therapy was also investigated using cell line. We found that the expression of CFL1 was lower in CML patients than that in healthy controls, and was significantly upregulated after imatinib therapy (p<0.05). CML patients with lower CFL1 achieved higher Major molecular response (MMR) rate after 6 months of imatinib therapy (p<0.05). Cofilin, P-cofilin and F-actin, especially branched F-actin were all upregulated after imatinib therapy. The lower CFL1 expression before treatment may predicts a better response to imatinib. Imatinib affects F-actin remodeling in CML patients by regulating CFL1 expression and activity.

18.
Article in English | MEDLINE | ID: mdl-38701878

ABSTRACT

BACKGROUND: Anhedonia, a core symptom of major depressive disorder (MDD), manifests in two forms: anticipatory and consummatory, reflecting a diminished capacity to anticipate or enjoy pleasurable activities. Prior studies suggest that brain-derived neurotrophic factor (BDNF) and interleukin-10 (IL-10) may play key roles in the emergence of anhedonia in MDD. The specific relationships between these biomarkers and the two forms of anhedonia remain unclear. This study investigated the potential links between BDNF, IL-10, and both forms of anhedonia in MDD patients. METHODS: This study included 43 participants diagnosed with MDD and 58 healthy controls. It involved detailed assessments of depression and anxiety levels, anticipatory and consummatory pleasure, cognitive functions, and a broad spectrum of plasma biomarkers, such as C-reactive protein, various interleukins, and BDNF. Using partial correlation, variables related to pleasant experiences were identified. Stepwise multiple linear regression analysis was applied to pinpoint the independent predictors of anhedonia in the MDD group. RESULTS: Demographically, both groups were comparable in terms of age, sex, body mass index, educational year, and marital status. Individuals with MDD displayed markedly reduced levels of anticipatory and consummatory pleasure, higher anxiety, and depression scores compared to healthy controls. Additionally, cognitive performance was notably poorer in the MDD group. These patients also had lower plasma diamine oxidase levels. Analysis linked anhedonia to impaired delayed memory. Regression results identified IL-10 and BDNF as independent predictors of anticipatory and consummatory anhedonia, respectively. CONCLUSION: These findings demonstrate that anticipatory and consummatory anhedonia are influenced by independent factors, thereby providing critical insights into the distinct neuroimmunological mechanisms that underlie various forms of anhedonia. Clinicl Trial Registration Number: NCT03790085.


Subject(s)
Anhedonia , Brain-Derived Neurotrophic Factor , Depressive Disorder, Major , Interleukin-10 , Humans , Depressive Disorder, Major/blood , Depressive Disorder, Major/psychology , Male , Anhedonia/physiology , Brain-Derived Neurotrophic Factor/blood , Female , Adult , Interleukin-10/blood , Middle Aged , Biomarkers/blood , Young Adult
19.
Article in English | MEDLINE | ID: mdl-38901758

ABSTRACT

BACKGROUND: Schizophrenia is a prevalent mental disorder, leading to severe disability. Currently, the absence of objective biomarkers hinders effective diagnosis. This study was conducted to explore the aberrant spontaneous brain activity and investigate the potential of abnormal brain indices as diagnostic biomarkers employing machine learning methods. METHODS: A total of sixty-one schizophrenia patients and seventy demographically matched healthy controls were enrolled in this study. The static indices of resting-state functional magnetic resonance imaging (rs-fMRI) including amplitude of low frequency fluctuations (ALFF), fractional ALFF (fALFF), regional homogeneity (ReHo), and degree centrality (DC) were calculated to evaluate spontaneous brain activity. Subsequently, a sliding-window method was then used to conduct temporal dynamic analysis. The comparison of static and dynamic rs-fMRI indices between the patient and control groups was conducted using a two-sample t-test. Finally, the machine learning analysis was applied to estimate the diagnostic value of abnormal indices of brain activity. RESULTS: Schizophrenia patients exhibited a significant increase ALFF value in inferior frontal gyrus, alongside significant decreases in fALFF values observed in left postcentral gyrus and right cerebellum posterior lobe. Pervasive aberrations in ReHo indices were observed among schizophrenia patients, particularly in frontal lobe and cerebellum. A noteworthy reduction in voxel-wise concordance of dynamic indices was observed across gray matter regions encompassing the bilateral frontal, parietal, occipital, temporal, and insular cortices. The classification analysis achieved the highest values for area under curve at 0.87 and accuracy at 81.28% when applying linear support vector machine and leveraging a combination of abnormal static and dynamic indices in the specified brain regions as features. CONCLUSIONS: The static and dynamic indices of brain activity exhibited as potential neuroimaging biomarkers for the diagnosis of schizophrenia.


Subject(s)
Brain , Machine Learning , Magnetic Resonance Imaging , Schizophrenia , Humans , Schizophrenia/diagnostic imaging , Schizophrenia/physiopathology , Magnetic Resonance Imaging/methods , Male , Female , Adult , Brain/diagnostic imaging , Brain/physiopathology , Rest/physiology , Young Adult , Brain Mapping/methods , Support Vector Machine
20.
Mol Neurobiol ; 59(10): 6307-6320, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35927544

ABSTRACT

Stroke, the leading cause of long-term disability worldwide, is caused by the blockage or hemorage of cerebral arteries. The resultant cerebral ischemia causes local neuronal death and brain injury. Histone deacetylase 9 (HDAC9) has been reported to be elevated in ischemic brain injury, but its mechanism in stroke is still enigmatic. The present study aimed to unveil the manner of regulation of HDAC9 expression and the effect of HDAC9 activation on neuronal function in cerebral ischemia. MicroRNAs (miRNAs) targeting HDAC9 were predicted utilizing bioinformatics analysis. We then constructed the oxygen glucose deprivation (OGD) cell model and the middle cerebral artery occlusion (MCAO) rat model, and elucidated the expression of CCCTC binding factor (CTCF)/miR-383-5p/HDAC9. Targeting between miR-383-5p and HDAC9 was verified by dual-luciferase reporter assay and RNAi. After conducting an overexpression/knockdown assay, we assessed neuronal impairment and brain injury. We found that CTCF inhibited miR-383-5p expression via its enrichment in the promoter region of miR-383-5p, whereas the miR-383-5p targeted and inhibited HDAC9 expression. In the OGD model and the MCAO model, we confirmed that elevation of HDAC9 regulated by the CTCF/miR-383-5p/HDAC9 pathway mediated apoptosis induced by endoplasmic reticulum stress, while reduction of HDAC9 alleviated apoptosis and the symptoms of cerebral infarction in MCAO rats. Thus, the CTCF/miR-383-5p/HDAC9 pathway may present a target for drug development against ischemic brain injury.


Subject(s)
Brain Injuries , Brain Ischemia , MicroRNAs , Reperfusion Injury , Stroke , Animals , Apoptosis , Brain Ischemia/genetics , Brain Ischemia/metabolism , CCCTC-Binding Factor , Glucose/metabolism , Histone Deacetylases , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Oxygen , Rats , Reperfusion Injury/metabolism , Stroke/metabolism , Transcription Factors
SELECTION OF CITATIONS
SEARCH DETAIL