Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Neuroimmunomodulation ; 29(1): 44-54, 2022.
Article in English | MEDLINE | ID: mdl-34518486

ABSTRACT

INTRODUCTION: Glioma is the most aggressive and malignant type of tumors among primary intracranial tumors. miR-433-3p has been verified to be correlated with the formation and progression of many types of cancers. METHODS: In this study, the effects of miR-433-3p and AJUBA on the proliferation, migration, and invasion of glioma and the molecular mechanisms were investigated. We analyzed bioinformatics databases and conducted cell biology experiments to determine that compared with adjacent tissue and normal cells, the expression level of miR-433-3p in glioma tissue and cells was lower, while the expression level of AJUBA was higher. Overexpressing miR-433-3p could significantly inhibit the proliferation, migration, and invasion of glioma cells and promote cell apoptosis. RESULTS: In addition, after overexpressing miR-433-3p and AJUBA, it was found that overexpressing AJUBA could attenuate the inhibitory effect of overexpressing miR-433-3p on the proliferation, migration, and invasion of glioma cells, which suggested that miR-433-3p regulated the biological function of glioma by downregulating AJUBA expression. CONCLUSION: These results proved that miR-433-3p could target to inhibit the expression of AJUBA, thus inhibiting the biological function and malignant progression of glioma.


Subject(s)
Brain Neoplasms , Glioma , LIM Domain Proteins , MicroRNAs , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Humans , LIM Domain Proteins/biosynthesis , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism
2.
Brain Res ; 1766: 147501, 2021 09 01.
Article in English | MEDLINE | ID: mdl-33915163

ABSTRACT

BACKGROUND: Circular RNA (circRNA) septin 9 (circSEPT9; hsa_circ_0005320) has been reported to be abnormally up-regulated in glioma. However, the exact role and working mechanism of circSEPT9 in glioma progression are barely known. METHODS: RNA and protein levels were measured by reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blot assay, respectively. Cell proliferation was assessed by 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, colony formation assay and flow cytometry. Cell apoptosis was evaluated by flow cytometry. Cell motility was analyzed by transwell assays. Cell glycolytic metabolism was analyzed using commercial kits. Dual-luciferase reporter assay, RNA-pull down assay and RNA immunoprecipitation (RIP) assay were conducted to verify the intermolecular interactions. Xenograft mice model was utilized to assess the role of circSEPT9 in vivo. RESULTS: CircSEPT9 was highly expressed in glioma tissues and cell lines. CircSEPT9 interference inhibited the proliferation, migration, invasion and glycolytic metabolism and triggered the apoptosis of glioma cells. MicroRNA-432-5p (miR-432-5p) was a target of circSEPT9, and circSEPT9 silencing-mediated effects in glioma cells were largely alleviated by the addition of anti-miR-432-5p. MiR-432-5p bound to the 3' untranslated region (3'UTR) of LIM and SH3 protein 1 (LASP1), and LASP1 overexpression largely overturned miR-432-5p-induced effects in glioma cells. CircSEPT9 up-regulated LASP1 expression by acting as miR-432-5p sponge. CircSEPT9 silencing suppressed xenograft tumor growth in vivo. CONCLUSION: CircSEPT9 exerted an oncogenic role to enhance the malignant behaviors of glioma cells by binding to miR-432-5p to induce LASP1 expression.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Brain Neoplasms/metabolism , Cytoskeletal Proteins/biosynthesis , Glioma/metabolism , LIM Domain Proteins/biosynthesis , MicroRNAs/biosynthesis , Septins/biosynthesis , Adaptor Proteins, Signal Transducing/genetics , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cytoskeletal Proteins/genetics , Glioma/genetics , Glioma/pathology , Humans , LIM Domain Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , RNA, Circular/biosynthesis , RNA, Circular/genetics , Septins/genetics , Xenograft Model Antitumor Assays/methods
3.
Medicine (Baltimore) ; 99(51): e23443, 2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33371071

ABSTRACT

BACKGOUND: This study aims to identify the expression of lipoma preferred partner (LPP) in Paget disease (PD) and to further understand the pathogenesis of PD. METHODS: Tissue microarray was used to evaluate the expression of LPP by immunohistochemistry in 40 PD patients. The results of LPP expression were combined with clinical and histopathological characteristics. Patient files were analyzed retrospectively. RESULTS: Twenty-one cases were mammary Paget disease (MPD) and 19 extramammary Paget disease (EMPD) involving the vulva, scrotum, and penis. LPP was expressed in PD and this expression was significantly greater in MPD versus EMPD (P = .031). The expression of LPP in MPD was significantly related with age (P = .009) and expression of Ki-67 (P = .011). No statistically significant differences were observed in LPP expression as related to sex, body location, and time of PD diagnosis. CONCLUSIONS: While LPP is expressed in both MPD and EMPD, the intensity of this expression is greater in MPD. LPP expression is positively correlated with Ki-67 and is more prevalent in middle-aged versus senior MPD patients. Further research is needed to determine its potential role in tumorigenesis and distribution.


Subject(s)
Cytoskeletal Proteins/biosynthesis , LIM Domain Proteins/biosynthesis , Neoplasms/pathology , Paget Disease, Extramammary/pathology , Paget's Disease, Mammary/pathology , Adult , Age Factors , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Female , Humans , Male , Middle Aged , Penile Neoplasms/pathology , Retrospective Studies , Testicular Neoplasms/pathology , Tissue Array Analysis , Vulvar Neoplasms/pathology
4.
Respir Res ; 21(1): 235, 2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32938459

ABSTRACT

BACKGROUND: Non-small cell lung cancer (NSCLC) is one of the major types of lung cancer, which is a prevalent human disease all over the world. LncRNA LINC01503 is a super-enhancer-driven long non-coding RNA that is dysregulated in several types of human cancer. However, its role in NSCLC remains unknown. METHODS: Thirty NSCLC patients were recruited between April 2012 and April 2016. Luciferase reporter assay, qRT-PCR, Cell Counting Kit-8 (CCK-8), Transwell migration assay, RNA pull-down assay, western blotting, 5-ethynyl-29-deoxyuridine (EdU) assays, and flow cytometry were utilized to characterize the roles and relationships among LINC01503, miR-342-3p, and LASP1 in NSCLC. The transplanted mouse model was built to examine their biological functions in vivo. RESULTS: We demonstrated that the expression of lncRNA LINC01503 and LIM and SH3 domain protein 1 (LASP1) were upregulated and miR-342-3p was downregulated in NSCLC samples and cell lines. Functional experiments revealed that inhibiting the expression of LINC01503 or over-expression of miR-342-3p inhibited NSCLC growth and metastasis both in vitro and in vivo. In addition, LINC01503 could bind to miR-342-3p and affect the expression of LASP1. CONCLUSION: These results provide a comprehensive analysis of the roles of LINC01503 as a competing endogenous RNA (ceRNA) in NSCLC progression.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Carcinoma, Non-Small-Cell Lung/metabolism , Cytoskeletal Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , LIM Domain Proteins/biosynthesis , Lung Neoplasms/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , A549 Cells , Adaptor Proteins, Signal Transducing/genetics , Adult , Aged , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/physiology , Cytoskeletal Proteins/genetics , Female , Humans , LIM Domain Proteins/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Middle Aged , RNA, Long Noncoding/genetics
5.
Transl Neurodegener ; 9(1): 32, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32746944

ABSTRACT

BACKGROUND: Diseases and disorders with a chronic neuroinflammatory component are often linked with changes in brain metabolism. Among neurodegenerative disorders, people living with human immunodeficiency virus (HIV) and Alzheimer's disease (AD) are particularly vulnerable to metabolic disturbances, but the mechanistic connections of inflammation, neurodegeneration and bioenergetic deficits in the central nervous system (CNS) are poorly defined. The particularly interesting new cysteine histidine-rich-protein (PINCH) is nearly undetectable in healthy mature neurons, but is robustly expressed in tauopathy-associated neurodegenerative diseases including HIV infection and AD. Although robust PINCH expression has been reported in neurons in the brains of patients with HIV and AD, the molecular mechanisms and cellular consequences of increased PINCH expression in CNS disease remain largely unknown. METHODS: We investigated the regulatory mechanisms responsible for PINCH protein-mediated changes in bioenergetics, mitochondrial subcellular localization and bioenergetic deficits in neurons exposed to physiological levels of TNFα or the HIV protein Tat. Changes in the PINCH-ILK-Parvin (PIP) complex association with cofilin and TESK1 were assessed to identify factors responsible for actin depolymerization and mitochondrial mislocalization. Lentiviral and pharmacological inhibition experiments were conducted to confirm PINCH specificity and to reinstate proper protein-protein complex communication. RESULTS: We identified MEF2A as the PINCH transcription factor in neuroinflammation and determined the biological consequences of increased PINCH in neurons. TNFα-mediated activation of MEF2A via increased cellular calcium induced PINCH, leading to disruption of the PIP ternary complex, cofilin activation by TESK1 inactivation, and actin depolymerization. The disruption of actin led to perinuclear mislocalization of mitochondria by destabilizing the kinesin-dependent mitochondrial transport machinery, resulting in impaired neuronal metabolism. Blocking TNFα-induced PINCH expression preserved mitochondrial localization and maintained metabolic functioning. CONCLUSIONS: This study reported for the first time the mechanistic and biological consequences of PINCH expression in CNS neurons in diseases with a chronic neuroinflammation component. Our findings point to the maintenance of PINCH at normal physiological levels as a potential new therapeutic target for neurodegenerative diseases with impaired metabolisms.


Subject(s)
Actins/metabolism , Adaptor Proteins, Signal Transducing/biosynthesis , Destrin/metabolism , Inflammation Mediators/metabolism , LIM Domain Proteins/biosynthesis , Mitochondria/metabolism , Neurons/metabolism , Actins/genetics , Adaptor Proteins, Signal Transducing/genetics , Brain/metabolism , Brain/pathology , Cells, Cultured , Destrin/genetics , Fetus , Gene Expression , Humans , LIM Domain Proteins/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mitochondria/pathology , Neurons/pathology
6.
Int Immunopharmacol ; 88: 106883, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32805696

ABSTRACT

Excessive inflammatory response-induced apoptosis and the degeneration of articular chondrocytes contribute to the development and progression of osteoarthritis. PDZ and LIM domain containing protein 2 (PDLIM2) has emerged as one of the pivotal regulators in orchestrating an inflammatory response through regulating the activity of transcription factor nuclear factor (NF)-κB. However, whether PDLIM2 participates in the articular chondrocyte-associated inflammatory response in osteoarthritis remains unknown. In the current study, we aimed to explore the biological function of PDLIM2 in lipopolysaccharide (LPS)-stimulated articular chondrocytes, an in vitro model of osteoarthritis. Herein, we found that PDLIM2 expression was significantly down-regulated in chondrocytes in response to LPS exposure. Functional experiments revealed that PDLIM2 overexpression increased the viability and decreased the apoptosis of chondrocytes following LPS treatment. Moreover, PDLIM2 overexpression attenuated LPS-induced degeneration of chondrocytes via the down-regulation of matrix metalloproteinase (MMP)-3 and MMP-13 and the up-regulation of COL2A1 and ACAN. In addition, the overexpression of PDLIM2 decreased LPS-induced production of interleukin (IL)-1ß, IL-6 and TNF-α. In contrast, depletion of PDLIM2 exhibited the opposite effect. Mechanism research elucidated that PDLIM2 repressed the activation of NF-κB signaling associated with the down-regulation of NF-κB p65 protein expression. PDLIM2 depletion-exacerbated LPS-induced injury was significantly reversed by NF-κB inhibition. Taken together, these results demonstrate that PDLIM2 overexpression attenuates LPS-induced injury of articular chondrocytes through the inactivation of NF-κB signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Chondrocytes/metabolism , Inflammation/metabolism , Joints/metabolism , LIM Domain Proteins/biosynthesis , NF-kappa B/metabolism , Adaptor Proteins, Signal Transducing/genetics , Aggrecans/metabolism , Animals , Apoptosis , Cell Line , Cell Survival/genetics , Chondrocytes/cytology , Chondrocytes/drug effects , Collagen Type II/metabolism , Cytokines/metabolism , Down-Regulation , LIM Domain Proteins/genetics , Lipopolysaccharides/pharmacology , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3/metabolism , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Signal Transduction
7.
Oxid Med Cell Longev ; 2020: 8189706, 2020.
Article in English | MEDLINE | ID: mdl-33414896

ABSTRACT

Intervertebral disc degeneration (IDD) is a prevalent disease characterized by low back pain. Increasing extracellular matrix (ECM) synthesis and decreasing nucleus pulposus cell (NPC) apoptosis are promising strategies to recover degenerated NP. LIM mineralization protein- (LMP-) 1 has anti-inflammatory potential and is a promising gene target for the treatment of NP degeneration. In this study, we measured the expression of LMP-1 in the NP of patients. Then, we constructed LMP-1-overexpressing NPCs using lentiviral vectors and investigated the effects of LMP-1 on cell proliferation, apoptosis, and ECM synthesis in NPCs. The results showed that LMP-1 was highly expressed in the NP of patients. LMP-1 overexpression significantly increased proliferation and decreased apoptosis in NPCs. The expression of collagen II and sulfated glycosaminoglycan (sGAG) in NPCs was also upregulated after LMP-1 was overexpressed. Moreover, we demonstrated that LMP-1 decreased apoptosis of NPCs by inhibiting NF-κB signaling activation. These findings suggest that LMP-1 plays an essential role in mediating apoptosis in NPCs by regulating NF-κB signaling and can be used as a gene target for the treatment of IDD.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Apoptosis , Cytoskeletal Proteins/biosynthesis , Intervertebral Disc Degeneration/metabolism , LIM Domain Proteins/biosynthesis , NF-kappa B/metabolism , Nucleus Pulposus/metabolism , Adult , Animals , Cell Proliferation , Collagen/metabolism , Extracellular Matrix/metabolism , Glycosaminoglycans/metabolism , Humans , Lentivirus , Magnetic Resonance Imaging , Male , Middle Aged , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction , Young Adult
8.
Diagn Pathol ; 14(1): 100, 2019 Sep 04.
Article in English | MEDLINE | ID: mdl-31484540

ABSTRACT

BACKGROUND: To evaluate the clinical utility of LIM Domain Only 2 (LMO2) negative and CD38 positive in diagnosis of Burkitt lymphoma (BL). METHODS: LMO2 and CD38 expression determined by immunohistochemistry in 75 BL, 12 High-grade B-cell lymphoma, NOS (HGBL,NOS) and 3 Burkitt-like lymphomas with the 11q aberration. RESULTS: The sensitivity and specificity of LMO2 negative for detecting BL were 98.67 and 100%, respectively; those of CD38 positive were 98.67 and 66.67%, respectively. The sensitivity and specificity of a combination of both for detecting BL were 97.33 and 100%, respectively. In our study, the combined LMO2 negative and CD38 positive results had a higher area under the curve than either LMO2 negative or CD38 positive alone. CONCLUSIONS: A combination of LMO2 negative and CD38 positive is useful for the diagnosis of Burkitt lymphoma.


Subject(s)
ADP-ribosyl Cyclase 1/analysis , Adaptor Proteins, Signal Transducing/analysis , Biomarkers, Tumor/analysis , Burkitt Lymphoma/diagnosis , LIM Domain Proteins/analysis , Membrane Glycoproteins/analysis , Proto-Oncogene Proteins/analysis , ADP-ribosyl Cyclase 1/biosynthesis , Adaptor Proteins, Signal Transducing/biosynthesis , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , Immunohistochemistry , LIM Domain Proteins/biosynthesis , Male , Membrane Glycoproteins/biosynthesis , Middle Aged , Proto-Oncogene Proteins/biosynthesis , Sensitivity and Specificity , Young Adult
9.
J Cell Biochem ; 120(8): 13651-13657, 2019 08.
Article in English | MEDLINE | ID: mdl-30945341

ABSTRACT

LASP2 (LIM and SH3 protein 2), a member of the LIM-protein subfamily of the nebulin group, was first identified as a splice variant of the nebulin gene. In the past, investigators mainly focused on the impact of LASP2 on cardiac diseases because of its identification in the myocardium. Recently, several studies have reported that LASP2 is associated with the progression of various cancers. However, there have been no investigations on the expression and function of LASP2 in pancreatic cancer (PC). In this study, we performed the quantitative real-time polymerase chain reaction and Western blot analysis to detect the expression of LASP2 in PC tissues and cell lines. PC cells were transfected with LASP2 overexpression plasmid or the negative control in the presence or absence of tumor growth factor-ß (TGF-ß). The transwell assays were used to measure the effects of LASP2 on PC cell migration and invasion. The protein expression of epithelial-mesenchymal transition (EMT) markers was detected using Western blot assay. Our results demonstrated that LASP2 was downregulated in PC tissues and cell lines. In addition, upregulation of LASP2 inhibited the PC cell migration and invasion. We also found that LASP2 upregulation reversed TGF-ß-induced EMT in PC cells. Taken together, we provided novel evidence supporting the tumor-suppressor role of LASP2 in PC and suggested it as a potential therapeutic target in PC treatment.


Subject(s)
Carrier Proteins/biosynthesis , Cell Movement , Cytoskeletal Proteins/biosynthesis , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , LIM Domain Proteins/biosynthesis , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation , Carrier Proteins/genetics , Cell Line, Tumor , Cytoskeletal Proteins/genetics , Humans , LIM Domain Proteins/genetics , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Transforming Growth Factor beta/genetics
10.
FEBS Lett ; 593(8): 777-787, 2019 04.
Article in English | MEDLINE | ID: mdl-30883710

ABSTRACT

Read-through or mutations of a stop codon resulting in translation of the 3'-UTR produce potentially toxic C-terminally extended proteins. However, quality control mechanisms for such proteins are poorly understood in mammalian cells. Here, a comprehensive analysis of the 3'-UTRs of genes associated with hereditary diseases identified novel arrest-inducing sequences in the 3'-UTRs of 23 genes that can repress the levels of their protein products. In silico analysis revealed that the hydrophobicity of the polypeptides encoded in the 3'-UTRs is correlated with arrest efficiency. These results provide new insight into quality control mechanisms mediated by 3'-UTRs to prevent the production of C-terminally extended cytotoxic proteins.


Subject(s)
3' Untranslated Regions/genetics , Protein Biosynthesis/genetics , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/chemistry , LIM Domain Proteins/biosynthesis , LIM Domain Proteins/chemistry , Muscle Proteins/biosynthesis , Muscle Proteins/chemistry , N-Acetylgalactosamine-4-Sulfatase/biosynthesis , N-Acetylgalactosamine-4-Sulfatase/chemistry , Quality Control
11.
Parasitol Res ; 118(4): 1147-1158, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30747294

ABSTRACT

Trypanosoma carassii is a flagellated bloodstream parasite of cyprinid fish with pathogenesis manifesting primarily as anemia in experimentally infected fish. This anemia is characterized by decreases in the number of circulating red blood cells (RBCs) during peak parasitemia. We examined changes in the key blood metrics and expression of genes known to be important in the regulation of erythropoiesis. Increasing parasitemia was strongly correlated with an overall decrease in the total number of circulating RBCs. Gene expression of key erythropoiesis regulators (EPO, EPOR, GATA1, Lmo2, and HIFα) and proinflammatory cytokines (IFNγ and TNFα) were measured and their expressions differed from those in fish made anemic by injections of phenylhydrazine (PHZ). Significant upregulation of pro-erythropoietic genes was observed in PHZ-induced anemia, but not during peak parasitic infection. Previously, we reported on functional characterization of goldfish erythropoietin (rgEPO) and its ability to induce survival and differentiation of erythroid progenitor cells in vitro. Treatment of goldfish during the infection with rgEPO reduced the severity of anemia but failed to fully prevent the onset of the anemic state in infected fish. Proinflammatory cytokines have been implicated in the suppression of erythropoiesis during trypanosomiasis, specifically the cytokines TNFα, IFNγ, and IL-1ß. Analysis of key proinflammatory cytokines revealed that mRNA levels of IFNγ and TNFα were upregulated in response to infection, but only TNFα increased in response to PHZ treatment. Synergistic activity of the proinflammatory cytokines may be required to sustain prolonged anemia. These findings provide insight into the relationship between T. carassii and host anemia and suggest that T. carassii may directly or indirectly suppress host erythropoiesis.


Subject(s)
Anemia/genetics , Cytokines/biosynthesis , Erythropoiesis/genetics , Gene Expression Regulation/genetics , Goldfish/parasitology , Parasitemia/pathology , Trypanosoma/classification , Anemia/parasitology , Animals , Erythrocyte Count , Erythropoietin/biosynthesis , GATA1 Transcription Factor/biosynthesis , Interferon-gamma/biosynthesis , LIM Domain Proteins/biosynthesis , Phenylhydrazines/pharmacology , RNA, Messenger/genetics , Receptors, Erythropoietin/biosynthesis , Trypanosomiasis/pathology , Tumor Necrosis Factor-alpha/biosynthesis
12.
Cell Rep ; 26(4): 1021-1032.e6, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30673598

ABSTRACT

Muscle LIM protein (MLP) has long been regarded as a muscle-specific protein. Here, we report that MLP expression is induced in adult rat retinal ganglion cells (RGCs) upon axotomy, and its expression is correlated with their ability to regenerate injured axons. Specific knockdown of MLP in RGCs compromises axon regeneration, while overexpression in vivo facilitates optic nerve regeneration and regrowth of sensory neurons without affecting neuronal survival. MLP accumulates in the cell body, the nucleus, and in axonal growth cones, which are significantly enlarged by its overexpression. Only the MLP fraction in growth cones is relevant for promoting axon extension. Additional data suggest that MLP acts as an actin cross-linker, thereby facilitating filopodia formation and increasing growth cone motility. Thus, MLP-mediated effects on actin could become a therapeutic strategy for promoting nerve repair.


Subject(s)
Central Nervous System/metabolism , Gene Expression Regulation , Growth Cones/metabolism , LIM Domain Proteins/biosynthesis , Muscle Proteins/biosynthesis , Nerve Regeneration , Optic Nerve/physiology , Retinal Ganglion Cells/metabolism , Vesicular Transport Proteins/biosynthesis , Animals , Axotomy , COS Cells , Central Nervous System/pathology , Chlorocebus aethiops , LIM Domain Proteins/genetics , Mice , Mice, Transgenic , Muscle Proteins/genetics , Rats , Retinal Ganglion Cells/pathology , Vesicular Transport Proteins/genetics
13.
Oncogene ; 38(1): 73-87, 2019 01.
Article in English | MEDLINE | ID: mdl-30082913

ABSTRACT

Primary glioblastoma is the most frequent human brain tumor in adults and is generally fatal due to tumor recurrence. We previously demonstrated that glioblastoma-initiating cells invade the subventricular zones and promote their radio-resistance in response to the local release of the CXCL12 chemokine. In this work, we show that the mitotic Aurora A kinase (AurA) is activated through the CXCL12-CXCR4 pathway in an ERK1/2-dependent manner. Moreover, the CXCL12-ERK1/2 signaling induces the expression of Ajuba, the main cofactor of AurA, which allows the auto-phosphorylation of AurA.We show that AurA contributes to glioblastoma cell survival, radio-resistance, self-renewal, and proliferation regardless of the exogenous stimulation with CXCL12. On the other hand, AurA triggers the CXCL12-mediated migration of glioblastoma cells in vitro as well as the invasion of the subventricular zone in xenograft experiments. Moreover, AurA regulates cytoskeletal proteins (i.e., Actin and Vimentin) and favors the pro-migratory activity of the Rho-GTPase CDC42 in response to CXCL12. Altogether, these results show that AurA, a well-known kinase of the mitotic machinery, may play alternative roles in human glioblastoma according to the CXCL12 concentration.


Subject(s)
Aurora Kinase A/physiology , Brain Neoplasms/enzymology , Chemokine CXCL12/physiology , Glioblastoma/enzymology , Neoplasm Proteins/physiology , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival , Chemokine CXCL12/pharmacology , Enzyme Activation , Glioblastoma/pathology , Heterografts , Humans , LIM Domain Proteins/biosynthesis , LIM Domain Proteins/genetics , Lateral Ventricles/pathology , MAP Kinase Signaling System , Mice , Neoplasm Invasiveness , Phosphorylation , Protein Processing, Post-Translational , Receptors, CXCR4/physiology , Signal Transduction
14.
Int J Biochem Cell Biol ; 106: 84-95, 2019 01.
Article in English | MEDLINE | ID: mdl-30453092

ABSTRACT

Large bone defects and bone loss after fractures remain significant challenges for orthopedic surgeons. Our study aims to find an available, applicable and biological treatment for bone regeneration overcoming the limitations in ESC/iPSC technology. We directly reprogrammed the mouse embryonic fibroblast (MEF) into osteoblast cells using different combinations of Yamanaka factors with human lim mineralization protein-3 (hLMP-3). LMP is an intracellular LIM-domain protein acting as an effective positive regulator of the osteoblast differentiation. After transduction, cells were cultured in osteogenic medium, and then examined for osteoblast formation. The expression of osteogenic markers (BMP2, Runx2 and Osterix) during reprogramming and in vitro mineralization assay revealed that the best reprogramming cocktail was (c-Myc - Oct4) with hLMP-3. In addition, both immunofluorescent staining and western blot analysis confirmed that osteocalcin (OCN) expression increased in the cells treated with the c-Myc/Oct4/hLMP3 cocktail than using hLMP-3 alone. Furthermore, this reprogramming cocktail showed efficient healing in an induced femoral bone defect in rat animal model one month after transplantation. In the present study, we reported for the first time the effect of combining Yamanaka factors with hLMP-3 to induce osteoblast cells from MEF both in vitro and in vivo.


Subject(s)
Cellular Reprogramming , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Intracellular Signaling Peptides and Proteins/biosynthesis , LIM Domain Proteins/biosynthesis , Osteoblasts/metabolism , Animals , Antigens, Differentiation/biosynthesis , Antigens, Differentiation/genetics , Cellular Reprogramming Techniques , Embryo, Mammalian/cytology , Fibroblasts/cytology , Humans , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , Mice , Osteoblasts/cytology
15.
Med Sci Monit ; 24: 7603-7613, 2018 Oct 25.
Article in English | MEDLINE | ID: mdl-30356034

ABSTRACT

BACKGROUND Extranodal NK/T cell lymphoma, nasal type (ENKTL-NT) is difficult to distinguish from nasal polyps and inverted papilloma, leading to its high misdiagnosis ratio. The aim of this study was to investigate its potential prognostic indicators. MATERIAL AND METHODS Kaplan-Meier method was used to calculate overall survival (OS) rate. Cox proportional hazards regression was used to analyze risk ratios (ORs) with 95% confidence intervals (CIs). RESULTS Nasal ala infiltration and nasal floor thickness >2.0 mm or nasal septum thickness >2.5 mm were potential prognostic factors for OS (p=0.0323 and 0.0072, respectively). Cox proportional-hazards regression indicated that high LMP1 expression and the nasal floor thickness >2.0 mm or nasal septum thickness >2.5 mm were the independent risk factors for poor OS of ENKTL-NT (HR=3.0655, p=0.028; HR=2.3650, p=0.0452, respectively). In the subgroup analysis, the OS rate was lower when the nasal floor thickness >2.0 mm or nasal septum thickness >2.5 mm in the patients who had high expression of LMP1 (p=0.0651), whereas high LMP1 expression increased the risk of worse prognostic outcome in patients with deep infiltration thickness. Thus, high LMP1 expression may contribute to the tissue invasion of ENKTL-NT. CONCLUSIONS Any patient with nasal ala soft-tissue invasion, nasal floor thickness >2.0 mm/nasal septum thickness >2.5 mm on CT imaging or high LMP1 expression should prompt immediate histopathologic diagnosis to rule out ENKTL-NT in clinical practice.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Cytoskeletal Proteins/biosynthesis , LIM Domain Proteins/biosynthesis , Lymphoma, Extranodal NK-T-Cell/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adult , Aged , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Female , Humans , Kaplan-Meier Estimate , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Lymphoma, Extranodal NK-T-Cell/diagnosis , Lymphoma, Extranodal NK-T-Cell/genetics , Male , Middle Aged , Neoplasm Invasiveness , Prognosis , Retrospective Studies , Survival Rate
16.
Int J Biochem Cell Biol ; 103: 14-24, 2018 10.
Article in English | MEDLINE | ID: mdl-30063986

ABSTRACT

Helicobacter pylori is the strongest known risk-factor for gastric cancer. However, its role in gastric cancer metastasis remains unclear. Previously we have reported that H. pylori promotes gastric cancer invasiveness by stabilizing the E3 ubiquitin ligase Siah2 which is mediated by Siah2 acetylation at Lys 139 (K139) residue. Here we identify that cell adhesion-related proteins testin (TES) and filamin-C (FLN-C) interact with Siah2 and get proteasomally degraded. The efficiency of TES and FLN-C degradation is significantly potentiated by K139-acetylated Siah2 (ac-K139 Siah2) in infected gastric cancer cells (GCCs). ac-Siah2-mediated downregulation of TES and FLN-C disrupts filopodia structures but promotes lamellipodia formation and enhances invasiveness and migration of infected GCCs. Since H. felis-infected mice as well as human gastric cancer biopsy samples also show high level of ac-K139 Siah2 and downregulated TES and FLN-C, we believe that acetylation of Siah2 is an important checkpoint that can be useful for therapeutic intervention.


Subject(s)
Cytoskeletal Proteins/biosynthesis , Down-Regulation , Filamins/biosynthesis , Gene Expression Regulation, Neoplastic , Helicobacter Infections/metabolism , Helicobacter pylori/metabolism , LIM Domain Proteins/biosynthesis , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Stomach Neoplasms , Ubiquitin-Protein Ligases/metabolism , Acetylation , Animals , Cell Line, Tumor , Humans , Mice , Neoplasm Invasiveness , RNA-Binding Proteins , Stomach Neoplasms/metabolism , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology
17.
Endocr Pathol ; 29(3): 222-230, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29560564

ABSTRACT

We previously described that LIM domain containing 2 (LIMD2) overexpression was closely correlated with metastatic process in papillary thyroid carcinoma (PTC). We here evaluated the expression of LIMD2 in a series of non-metastatic and metastatic PTC and their matched lymph node metastases via immunohistochemistry. LIMD2 was expressed in 74 (81%) of primary PTC and 35 (95%) of lymph node metastases. Sub-analysis performed in 37 matched samples demonstrated that in four cases, LIMD2 is expressed in lymph node metastases, while it is not expressed in primary tumors. Moreover, in eight cases, the staining intensity of LIMD2 was stronger in the patient-matched lymph node metastases than in the primary tumors. Next, the expression of LIMD2 was correlated with clinical pathological parameters and BRAF V600E and RET/PTC mutational status. The expression of LIMD2 in primary tumors was correlated with the presence of BRAF V600E mutation (P = 0.0338). Western blot analysis in thyroid cell lines demonstrated that LIMD2 is expressed in two PTC cell lines, while it is not expressed in normal thyroid and follicular thyroid carcinoma cell lines. Importantly, its expression was higher in a PTC cell line that harbors BRAF V600E mutation than in a PTC cell line that harbors RET/PTC1. The available genomic profiling data generated by The Cancer Genome Atlas Research Network confirmed that LIMD2 expression is higher in BRAF-like PTC samples. Our data suggest that LIMD2 may play an important role in the metastatic process of PTC, predominantly in BRAF V600E-positive tumors.


Subject(s)
Biomarkers, Tumor/analysis , LIM Domain Proteins/biosynthesis , Lymphatic Metastasis/pathology , Thyroid Cancer, Papillary/pathology , Adult , Aged , Female , Humans , Male , Middle Aged , Mutation , Proto-Oncogene Proteins B-raf/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Cancer, Papillary/metabolism , Up-Regulation
18.
Int J Oncol ; 52(1): 179-188, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29138807

ABSTRACT

Lung cancer accounts for most cancer-related deaths worldwide. However, the underlying mechanism by which it mediates the progression of lung cancer remains unclear. Expression of LASP-1 (LIM and SH3 protein 1) was evaluated in lung cancer tissues and tumor-adjacent normal tissues using immunohistochemistry and western blotting. Functional studies have shown that siRNA-mediated silencing of LASP-1 in human lung cancer cells and reduced cell proliferation, migration, and invasion. Flow cytometry and immunofluorescence staining also revealed that rate of cell apoptosis was increased after knockdown of expression of LASP-1, thereby suggesting that LASP-1 may function as an oncogene during lung cancer progression. SOX9 is an important transcription factor, which is involved in the development of several types of human cancer. Further analysis has showed the presence of a consensus-binding site of SOX9 in the promoter region of LASP-1. Mechanistic investigations showed that LASP-1 was transcriptionally activated by SOX9. Through luciferase reporter and ChIP assays, we demonstrated that LASP-1 was a direct target gene of sex determining region Y-box 9 (SOX9). Knockdown of SOX9 expression by RNA interference reduces cell proliferation and induces apoptosis of lung cancer cells, which was consistent with the results obtained from silencing the expression of LASP-1 in NCI­H1650 cells. Together, these findings indicated that LASP-1, as a downstream target of SOX9, may act as a novel biomarker for lung cancer and plays an important role in cell proliferation, migration, and invasion.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cytoskeletal Proteins/genetics , LIM Domain Proteins/genetics , Lung Neoplasms/genetics , SOX9 Transcription Factor/genetics , A549 Cells , Adaptor Proteins, Signal Transducing/biosynthesis , Aged , Aged, 80 and over , Cell Line, Tumor , Cytoskeletal Proteins/biosynthesis , Disease Progression , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , LIM Domain Proteins/biosynthesis , Lung Neoplasms/metabolism , Male , Middle Aged , SOX9 Transcription Factor/metabolism , Transcription, Genetic
19.
Tumour Biol ; 39(6): 1010428317705757, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28621232

ABSTRACT

Although usually referred to as a structural actin-binding protein, LIM and SH3 domain-containing protein may actually be dynamically involved in the control of a wide spectrum of cellular processes, by virtue of its interaction with several molecular partners. Alongside being ubiquitously expressed in physiological conditions, LIM and SH3 domain-containing protein is overexpressed in a growing number of human cancers, in which it may actively contribute to their aggressiveness by promoting cell proliferation and migration. In view of the recent findings, implicating the protein in cancer progression, we discuss here the most relevant discoveries highlighting the role of this versatile protein in various human tumors. The correlation between LIM and SH3 domain-containing protein expression levels in cancer and the poor outcome and metastatic behavior of tumors denotes the clinical significance of this protein and hints its potential value as a new cancer prognostic or even diagnostic biomarker. This may be decisive not only to optimize existing pharmacological regimes but also to delineate novel, more efficacious therapeutic and/or preventive approaches.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Cytoskeletal Proteins/genetics , LIM Domain Proteins/genetics , Neoplasms/genetics , Adaptor Proteins, Signal Transducing/biosynthesis , Biomarkers, Tumor/biosynthesis , Cell Proliferation/genetics , Cytoskeletal Proteins/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , LIM Domain Proteins/biosynthesis , Neoplasms/pathology , Prognosis
20.
Leukemia ; 31(10): 2057-2064, 2017 10.
Article in English | MEDLINE | ID: mdl-28260788

ABSTRACT

Oncogenic driver mutations are those that provide a proliferative or survival advantage to neoplastic cells, resulting in clonal selection. Although most cancer-causing mutations have been detected in the protein-coding regions of the cancer genome; driver mutations have recently also been discovered within noncoding genomic sequences. Thus, a current challenge is to gain precise understanding of how these unique genomic elements function in cancer pathogenesis, while clarifying mechanisms of gene regulation and identifying new targets for therapeutic intervention. Here we report a C-to-T single nucleotide transition that occurs as a somatic mutation in noncoding sequences 4 kb upstream of the transcriptional start site of the LMO1 oncogene in primary samples from patients with T-cell acute lymphoblastic leukaemia. This single nucleotide alteration conforms to an APOBEC-like cytidine deaminase mutational signature, and generates a new binding site for the MYB transcription factor, leading to the formation of an aberrant transcriptional enhancer complex that drives high levels of expression of the LMO1 oncogene. Since APOBEC-signature mutations are common in a broad spectrum of human cancers, we suggest that noncoding nucleotide transitions such as the one described here may activate potent oncogenic enhancers not only in T-lymphoid cells but in other cell lineages as well.


Subject(s)
APOBEC Deaminases/metabolism , DNA-Binding Proteins/biosynthesis , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Leukemic/genetics , LIM Domain Proteins/biosynthesis , Neoplasm Proteins/biosynthesis , Point Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcription Factors/biosynthesis , Transcriptome , 5' Untranslated Regions/genetics , Base Sequence , Binding Sites , Cell Line, Tumor , Child , Chromatin Immunoprecipitation , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , Genes, myb , Humans , Jurkat Cells , LIM Domain Proteins/genetics , Neoplasm Proteins/genetics , Polymorphism, Single Nucleotide , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Proteins c-myb/antagonists & inhibitors , Proto-Oncogene Proteins c-myb/genetics , Proto-Oncogene Proteins c-myb/metabolism , RNA Interference , RNA, Small Interfering/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...