Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
J Biol Chem ; 300(5): 107286, 2024 May.
Article in English | MEDLINE | ID: mdl-38636657

ABSTRACT

Hepatitis C virus (HCV) infection is tightly connected to the lipid metabolism with lipid droplets (LDs) serving as assembly sites for progeny virions. A previous LD proteome analysis identified annexin A3 (ANXA3) as an important HCV host factor that is enriched at LDs in infected cells and required for HCV morphogenesis. To further characterize ANXA3 function in HCV, we performed proximity labeling using ANXA3-BioID2 as bait in HCV-infected cells. Two of the top proteins identified proximal to ANXA3 during HCV infection were the La-related protein 1 (LARP1) and the ADP ribosylation factor-like protein 8B (ARL8B), both of which have been previously described to act in HCV particle production. In follow-up experiments, ARL8B functioned as a pro-viral HCV host factor without localizing to LDs and thus likely independent of ANXA3. In contrast, LARP1 interacts with HCV core protein in an RNA-dependent manner and is translocated to LDs by core protein. Knockdown of LARP1 decreased HCV spreading without altering HCV RNA replication or viral titers. Unexpectedly, entry of HCV particles and E1/E2-pseudotyped lentiviral particles was reduced by LARP1 depletion, whereas particle production was not altered. Using a recombinant vesicular stomatitis virus (VSV)ΔG entry assay, we showed that LARP1 depletion also decreased entry of VSV with VSV, MERS, and CHIKV glycoproteins. Therefore, our data expand the role of LARP1 as an HCV host factor that is most prominently involved in the early steps of infection, likely contributing to endocytosis of viral particles through the pleiotropic effect LARP1 has on the cellular translatome.


Subject(s)
Annexin A3 , Hepacivirus , Hepatitis C , SS-B Antigen , Virus Internalization , Humans , Annexin A3/metabolism , Annexin A3/genetics , Autoantigens/metabolism , Autoantigens/genetics , HEK293 Cells , Hepacivirus/metabolism , Hepacivirus/physiology , Hepatitis C/metabolism , Hepatitis C/virology , Hepatitis C/genetics , Host-Pathogen Interactions , Lipid Droplets/metabolism , Lipid Droplets/virology , Ribonucleoproteins/metabolism , Ribonucleoproteins/genetics , Viral Core Proteins/metabolism , Viral Core Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Envelope Proteins/genetics
2.
Cancer Immunol Res ; 12(5): 614-630, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38393971

ABSTRACT

Tumor-associated macrophages (TAM) induce immunosuppression in laryngeal squamous cell carcinoma (LSCC). The interaction between LSCC cells and TAMs affects the progression of laryngeal cancer through exosomes, but the underlying molecular mechanism remains unclear. Proteomics analysis of TAMs isolated from human laryngeal tumor tissues obtained from patients with confirmed lymphatic metastasis revealed an upregulation of annexin A3 (ANXA3). In TAMs, ANXA3 promoted macrophages to polarize to an M2-like phenotype by activating the AKT-GSK3ß-ß-catenin pathway. In addition, ANXA3-rich exosomes derived from TAMs inhibited ferroptosis in laryngeal cancer cells through an ATF2-CHAC1 axis, and this process was associated with lymphatic metastasis. Mechanistically, ANXA3 in exosomes inhibited the ubiquitination of ATF2, whereas ATF2 acted as a transcription factor to regulate the expression of CHAC1, thus inhibiting ferroptosis in LSCC cells. These data indicate that abnormal ANXA3 expression can drive TAM reprogramming and promote an immunosuppressive microenvironment in LSCC. Meanwhile, ANXA3-rich exosomes inhibit ferroptosis of LSCC cells and promote lymphatic metastasis, thus promoting tumor progression.


Subject(s)
Annexin A3 , Exosomes , Ferroptosis , Laryngeal Neoplasms , Tumor-Associated Macrophages , Animals , Humans , Male , Mice , Annexin A3/metabolism , Cell Line, Tumor , Exosomes/metabolism , Laryngeal Neoplasms/pathology , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/immunology , Lymphatic Metastasis/immunology , Lymphatic Metastasis/pathology , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/immunology , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology
3.
Mol Immunol ; 167: 25-33, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38310670

ABSTRACT

Acute lung injury (ALI) is a prevailing and deadly complication of sepsis coupled with increasing incidence and fatality rate. Annexin A3 (ANXA3) has been unraveled to be upregulated during sepsis. This study purposed to assess the role and the mechanism of ANXA3 in sepsis-induced ALI. After the construction of mouse model of sepsis, the pathological changes of mice lung tissues were estimated by H&E staining. ANXA3 expression in mice lung tissues and serum was examined. The degree of pulmonary edema and the levels of inflammatory factors in bronchoalveolar lavage fluid (BALF) were analyzed. In lipopolysaccharide (LPS)-induced mouse ALI model in vitro, CCK-8 assay measured cell viability and flow cytometry analysis detected cell apoptosis. Besides, ELISA assay detected the release of inflammatory cytokines. Western blot analyzed the expression of proteins associated with inflammation, apoptosis and extracellular-signal-regulated kinase (ERK)/ETS-like gene 1 (ELK1) signaling. Results revealed that ANXA3 was overexpressed in the lung tissues and serum of septic mice. Following the knockdown of ANXA3, sepsis-induced lung injury was alleviated, manifested as reduced lung edema, decreased inflammatory cell infiltration and inhibited cell apoptosis. Additionally, ANXA3 silence blocked ERK/ELK1 signaling both in sepsis mouse models and in vitro model of ALI induced by lipopolysaccharide (LPS). Moreover, the inhibitory effects of ANXA3 silencing on ERK/ELK1 signaling activation, the viability damage, inflammation and apoptosis in LPS-induced mouse ALI model in vitro were partially reversed by ERK activator. Collectively, depletion of ANXA3 exerted suppressive effects on the inflammation and apoptosis in sepsis-induced ALI through blocking ERK/ELK1 signaling.


Subject(s)
Acute Lung Injury , Sepsis , Animals , Mice , Acute Lung Injury/pathology , Annexin A3/metabolism , Apoptosis , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Inflammation/pathology , Lipopolysaccharides/pharmacology , Lung/metabolism , Sepsis/metabolism
4.
Eur J Med Res ; 28(1): 596, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38102696

ABSTRACT

BACKGROUND: Pyroptosis plays an important role in the pathological process of ischemic stroke (IS). However, the exact mechanism of pyroptosis remains unclear. This paper aims to reveal the key molecular markers associated with pyroptosis in IS. METHODS: We used random forest learning, gene set variation analysis, and Pearson correlation analysis to screen for biomarkers associated with pyroptosis in IS. Middle cerebral artery occlusion/reperfusion (MCAO/R) and oxygen and glucose deprivation/reoxygenation (OGD/R) models were constructed in vitro and in vivo. Cells were transfected with an Annexin A3 silencing (si-ANXA3) plasmid to observe the effects of ANXA3 on OGD/R + lipopolysaccharides (LPS)-induced pyroptosis. qRT‒PCR and western blotting were used to detect the expression of potential biomarkers and pyroptotic pathways. RESULTS: Samples from a total of 170 IS patients and 109 healthy individuals were obtained from 5 gene expression omnibus databases. Thirty important genes were analyzed by random forest learning from the differentially expressed genes. Then, we investigated the relationship between the above genes and the pyroptosis score, obtaining three potential biomarkers (ANXA3, ANKRD22, ADM). ANXA3 and ADM were upregulated in the MCAO/R model, and the fold difference in ANXA3 expression was greater. Pyroptosis-related factors (NLRP3, NLRC4, AIM2, GSDMD-N, caspase-8, pro-caspase-1, cleaved caspase-1, IL-1ß, and IL-18) were upregulated in the MCAO/R model. Silencing ANXA3 alleviated the expression of pyroptosis-related factors (NLRC4, AIM2, GSDMD-N, caspase-8, pro-caspase-1, cleaved caspase-1, and IL-18) induced by OGD/R + LPS or MCAO/R. CONCLUSION: This study identified ANXA3 as a possible pyroptosis-related gene marker in IS through bioinformatics and experiments. ANXA3 could inhibit pyroptosis through the NLRC4/AIM2 axis.


Subject(s)
Ischemic Stroke , Reperfusion Injury , Humans , Pyroptosis/genetics , Interleukin-18/metabolism , Interleukin-18/pharmacology , Caspase 1/metabolism , Caspase 1/pharmacology , Caspase 8/metabolism , Caspase 8/pharmacology , Ischemic Stroke/genetics , Lipopolysaccharides/pharmacology , Biomarkers , Reperfusion Injury/genetics , Reperfusion Injury/metabolism , Annexin A3/genetics , Annexin A3/metabolism , Annexin A3/pharmacology
5.
Genes Genomics ; 45(12): 1575-1586, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37843781

ABSTRACT

BACKGROUND: Cervical cancer, as one of the most common cancers in women, remains a major health threat worldwide. Annexin A3 (ANXA3), a component of the annexin family, is upregulated in numerous cancers, with no explicit role in cervical cancer. OBJECTIVE: This study aims to investigate the function of ANXA3 in cervical cancer. METHODS: Differential expression genes between the cervical cancer tissues of patients and the controls were analyzed in The Cancer Genome Atlas (TCGA) and Gene Expression Profiling Interactive Analysis (GEPIA) database. Using transfection approaches to either upregulate or downregulate ANXA3, its role in cell proliferation and chemosensitivity of human cervical cancer cell lines (HeLa and C33A) was evaluated. Furthermore, the binding activity between YAP1 and ANXA3 was also explored. RESULTS: Genomics analysis indicated that differential genes were mostly associated with cell cycle progression and DNA replication. ANXA3 was highly expressed in the cervical cancer tissues and closely linked to malignancy degree. Knockdown of ANXA3 in cervical cancer cells inhibited cell cycle progression. A similar result was observed in the reduction of cyclin D, CDK4, cyclin E, and CDK2 in cervical cancer cells with ANXA3 silencing. Cervical cancer cells obtained high sensitivity to cisplatin (DDP) when ANXA3 was downregulated. Conversely, these capabilities were the opposite in cervical cancer cells overexpressing ANXA3. Furthermore, the expression levels of ANXA3 and YAP1 were positively correlated. YAP1 upregulation was positively connected with malignant behaviors, which were reversed by ANXA3 downregulation. CONCLUSION: In light of our findings, targeting ANXA3 expressed in cervical cancer might contribute to more potential therapeutic strategies.


Subject(s)
Annexin A3 , Uterine Cervical Neoplasms , Female , Humans , Annexin A3/genetics , Annexin A3/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Drug Resistance, Neoplasm/genetics , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics
6.
Bone ; 172: 116758, 2023 07.
Article in English | MEDLINE | ID: mdl-37030499

ABSTRACT

Annexin A3 (ANXA3), a member of Annexin family, is reported to mediate membrane transport and cancer development. However, the effect of ANXA3 on osteoclast formation and bone metabolism is still unclear. In this study, we found that knockdown of ANXA3 can significantly inhibit receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation through NF-κB signaling. ANXA3 downregulation abrogated the expression of osteoclast-specific genes, including Acp5, Mmp9 and Ctsk in osteoclast precursors. Moreover, lentiviral of shRNA against ANXA3 reversed the bone loss in osteoporosis using ovariectomized mice model. Mechanistically, we found that ANXA3 directly bound to RANK and TRAF6 to accelerate osteoclast differentiation by promoting their transcription and limiting degradation. In conclusion, we propose a fundamentally novel RANK-ANXA3-TRAF6 complex to effectively modulate the formation and differentiation of osteoclast to manipulate bone metabolism. The ANXA3-targeted therapeutic strategy may provide new insight for bone degrading-related diseases prevention and treatment.


Subject(s)
Bone Resorption , Osteoclasts , Mice , Animals , Osteoclasts/metabolism , TNF Receptor-Associated Factor 6/metabolism , Annexin A3/metabolism , Annexin A3/pharmacology , Bone and Bones/metabolism , Signal Transduction , NF-kappa B/metabolism , RANK Ligand/metabolism , Cell Differentiation , Bone Resorption/metabolism , Osteogenesis
7.
Stem Cell Reports ; 17(8): 1842-1858, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35868309

ABSTRACT

Exosomes play a critical role in intracellular communication. The biogenesis and function of exosomes are regulated by multiple biochemical factors. In the present study, we find that mechanical force promotes the biogenesis of exosomes derived from periodontal ligament stem cells (PDLSCs) and alters the exosomal proteome profile to induce osteoclastic differentiation. Mechanistically, mechanical force increases the level of exosomal proteins, especially annexin A3 (ANXA3), which facilitates exosome internalization to activate extracellular signal-regulated kinase (ERK), thus inducing osteoclast differentiation. Moreover, the infusion of exosomes derived from PDLSCs into mice promotes mechanical force-induced tooth movement and increases osteoclasts in the periodontal ligament. Collectively, this study demonstrates that mechanical force treatment promotes the biogenesis of exosomes from PDLSCs and increases exosomal protein ANXA3 to facilitate exosome internalization, which activates ERK phosphorylation, thus inducing osteoclast differentiation. Our findings shed light on new mechanisms for how mechanical force regulates the biology of exosomes and bone metabolism.


Subject(s)
Annexin A3 , Periodontal Ligament , Animals , Annexin A3/metabolism , Cell Differentiation/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Osteoclasts , Osteogenesis/physiology , Stem Cells/metabolism
8.
Exp Cell Res ; 418(2): 113292, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35850266

ABSTRACT

Radioresistance currently poses a significant challenge to successful disease control of nasopharyngeal carcinoma (NPC). We previously uncovered that annexin A3 (ANXA3), a calcium-dependent phospholipid binding protein, is underexpressed in radioresistant NPC cells and mouse xenografts. This study aims to further unravel the mechanistic basis underlying ANXA3-mediated radioresistance in NPC. We show that either innate ANXA3 downregulation or short hairpin RNA(shRNA)-based knockdown of ANXA3 confers resistance to ionizing radiation (IR) in NPC both in vitro and in mouse xenograft models in vivo, whereas radiosensitization was observed when ANXA3 was ectopically expressed. Mechanistically, ANXA3 knockdown dramatically enhances IR-induced epidermal growth factor receptor (EGFR) phosphorylation and nuclear translocation, leading to increased post-IR phosphorylation of DNA-dependent protein kinase catalytic subunit (DNA-PKcs) concomitant with markedly accelerated DNA DSB repair. In addition, pretreatment with cetuximab efficiently abrogated the radioresistant phenotype of ANXA3-low cells as well as the ANXA3 knockdown-induced post-IR EGFR nuclear accumulation, suggesting that EGFR is an essential mediator for ANXA3 depletion-mediated radioprotection in NPC. Collectively, this work reveals for the first time a critical role of ANXA3 in radiation survival and DNA repair mechanism of NPC and provides mechanistic evidence to support ANXA3 as a potential therapeutic target to improve radiocurability for NPC.


Subject(s)
Annexin A3 , Nasopharyngeal Neoplasms , Animals , Annexin A3/genetics , Annexin A3/metabolism , Cell Line, Tumor , DNA , Down-Regulation/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Carcinoma/radiotherapy , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/radiotherapy , RNA, Small Interfering , Radiation Tolerance/genetics , Radiation, Ionizing , Xenograft Model Antitumor Assays
9.
Front Immunol ; 13: 861459, 2022.
Article in English | MEDLINE | ID: mdl-35464477

ABSTRACT

Introduction: The specific pathogenesis of ankylosing spondylitis (AS) remains unclear, and our study aimed to investigate the possible pathogenesis of AS. Materials and Methods: Two datasets were downloaded from the GEO database to perform differentially expressed gene analysis, GO enrichment analysis, KEGG pathway analysis, DO enrichment analysis, GSEA analysis of differentially expressed genes, and construction of diagnostic genes using SVM and WGCNA along with Hypoxia-related genes. Also, drug sensitivity analysis was performed on diagnostic genes. To identify the differentially expressed immune genes in the AS and control groups, we analyzed the composition of immune cells between them. Then, we examined differentially expressed genes in three AS interspinous ligament specimens and three Degenerative lumbar spine specimens using high-throughput sequencing while the immune cells were examined using the neutrophil count data from routine blood tests of 1770 HLA-B27-positive samples and 7939 HLA-B27-negative samples. To assess the relationship between ANXA3 and SORL1 and disease activity, we took the neutrophil counts of the first 50 patients with above-average BASDAI scores and the last 50 patients with below-average BASDAI scores for statistical analysis. We used immunohistochemistry to verify the expression of ANXA3 and SORL1 in AS and in controls. Results: ANXA3 and SORL1 were identified as new diagnostic genes for AS. These two genes showed a significant differential expression between AS and controls, along with showing a significant positive correlation with the neutrophil count. The results of high-throughput sequencing verified that these two gene deletions were indeed differentially expressed in AS versus controls. Data from a total of 9707 routine blood tests showed that the neutrophil count was significantly higher in AS patients than in controls (p < 0.001). Patients with AS with a high BASDAI score had a much higher neutrophil count than those with a low score, and the difference was statistically significant (p < 0.001). The results of immunohistochemistry showed that the expression of ANXA3 and SORL1 in AS was significantly higher than that in the control group. Conclusion: Upregulated of ANXA3, SORL1, and neutrophils may be a key factor in the progression of Ankylosing spondylitis.


Subject(s)
Spondylitis, Ankylosing , Annexin A3/metabolism , HLA-B27 Antigen/genetics , Humans , LDL-Receptor Related Proteins , Leukocyte Count , Membrane Transport Proteins , Neutrophils/metabolism , Spondylitis, Ankylosing/diagnosis
10.
Am J Pathol ; 192(3): 426-440, 2022 03.
Article in English | MEDLINE | ID: mdl-34896071

ABSTRACT

Cerebral amyloid angiopathy (CAA), characterized by cerebral vascular amyloid accumulation, neuroinflammation, microbleeds, and white matter (WM) degeneration, is a common comorbidity in Alzheimer disease and a prominent contributor to vascular cognitive impairment and dementia. WM loss was recently reported in the corpus callosum (CC) in the rTg-DI rat model of CAA. The current study shows that the CC exhibits a much lower CAA burden compared with the adjacent cortex. Sequential Window Acquisition of All Theoretical Mass Spectra tandem mass spectrometry was used to show specific proteomic changes in the CC with emerging WM loss and compare them with the proteome of adjacent cortical tissue in rTg-DI rats. In the CC, annexin A3, heat shock protein ß1, and cystatin C were elevated at 4 months (M) before WM loss and at 12M with evident WM loss. Although annexin A3 and cystatin C were also enhanced in the cortex at 12M, annexin A5 and the leukodystrophy-associated astrocyte proteins megalencephalic leukoencephalopathy with subcortical cysts 1 and GlialCAM were distinctly elevated in the CC. Pathway analysis indicated neurodegeneration of axons, reflected by reduced expression of myelin and neurofilament proteins, was common to the CC and cortex; activation of Tgf-ß1 and F2/thrombin was restricted to the CC. This study provides new insights into the proteomic changes that accompany WM loss in the CC of rTg-DI rats.


Subject(s)
Cerebral Amyloid Angiopathy , White Matter , Animals , Annexin A3/metabolism , Brain/metabolism , Cerebral Amyloid Angiopathy/metabolism , Cystatin C/metabolism , Proteomics , Rats , White Matter/metabolism
11.
J Adv Res ; 30: 85-102, 2021 05.
Article in English | MEDLINE | ID: mdl-34026289

ABSTRACT

Introduction: As a member of annexin family proteins, annexin A3 (ANXA3) has 36-kDa and 33-kDa isoforms. ANXA3 plays crucial roles in the tumorigenesis, aggressiveness and drug-resistance of cancers. However, previous studies mainly focused on the role of total ANXA3 in cancers without distinguishing the distinction between the two isoforms, the role of 33-kDa ANXA3 in cancer remains unclear. Objectives: Current work aimed to investigate the function and regulation mechanism of 33-kDa ANXA3 in hepatocarcinoma. Methods: The expressions of ANXA3, CRKL, Rac1, c-Myc and pAkt were analyzed in hepatocarcinoma specimens by Western blotting. The biological function of 33-kDa ANXA3 in the growth, metastasis, apoptosis, angiogenesis, chemoresistance of hepatocarcinoma cells with the underlying molecular mechanism were investigated using gain-of-function strategy in vitro or in vivo. Results: 33-kDa ANXA3 was remarkably upregulated in tumor tissues compared with corresponding normal liver tissues of hepatocarcinoma patients. Its stable knockdown decreased the in vivo tumor growing velocity and malignancy of hepatocarcinoma HepG2 cells transplanted in nude mice. The in vitro experimental results indicated 33-kDa ANXA3 knockdown suppressed the proliferation, colony forming, migration and invasion abilities of HepG2 cells through downregulating CRKL, Rap1b, Rac1, pMEK, pERK2 and c-Myc in ERK pathway; inhibited angiogenesisability of HepG2 cells through inactivating PI3K/Akt-HIF pathway; induced apoptosis and enhanced chemoresistance of HepG2 cells through increasing Bax/decreasing Bcl-2 expressions and inactivating caspase 9/caspase 3 in intrinsic apoptosis pathway. Accordingly, CRKL, Rac1, c-Myc and pAkt were also upregulated in hepatocarcinoma patients ' tumor tissues compared with corresponding normal liver tissues. Conclusions: The overexpression of 33-kDa ANXA3 is involved in the clinical progression of hepatocarcinoma and in the malignancy, angiogenesis and apoptosis of hepatocarcinoma cells. It is of potential use in hepatocarcinoma diagnosis and treatment.


Subject(s)
Annexin A3/metabolism , Apoptosis , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Animals , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/pathology , Cell Movement , Cell Proliferation , Drug Resistance, Neoplasm , Female , Hep G2 Cells , Humans , Liver Neoplasms/pathology , MAP Kinase Signaling System , Male , Mice , Mice, Nude , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/metabolism
12.
Vet Microbiol ; 256: 109042, 2021 May.
Article in English | MEDLINE | ID: mdl-33819840

ABSTRACT

Bovine leukemia virus (BLV) is the causative agent of enzootic bovine leukosis (EBL) and can be classified into two types based on the amino acid at position 233 in Tax protein, which probably plays crucial roles in leukemogenesis. We previously revealed that L233-Tax-expressing cells secreted chemoattractants for endothelial cells and formed significantly larger tumors accompanying neovascularization than P233-Tax-expressing cells in athymic mice. In the present study, comparative proteomic analysis of the culture medium of Tax-expressing cells revealed that annexin A3 and probably extracellular matrix protein 1 served as chemoattractants. Conversely, L233-Tax-expressing cells were impaired in the secretion of collagen alpha-1 (I) chain precursor, which participates in tissue tension homeostasis, leading to tumor mass development. The analysis also demonstrated that both L233-Tax- and P233-Tax-expressing cells had deficits in the secretion of potentially antiangiogenic molecules, including pigment epithelium-derived factor and collagen alpha-1 (VIII) chain, and they produced complement component 3, which might participate in tumor cell proliferation, metastasis, and immune evasion. These findings provided novel insights into prognostication of EBL and the function of Tax in leukemogenesis induced by BLV.


Subject(s)
Annexin A3/metabolism , Collagen Type I/metabolism , Enzootic Bovine Leukosis/virology , Gene Products, tax/metabolism , Leukemia Virus, Bovine/genetics , Amino Acid Substitution , Animals , Cattle , Cell Line , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/virology , Fibroblasts/metabolism , Fibroblasts/virology , Gene Products, tax/genetics , Mutation , Proteomics , Rats
13.
Acta Neurobiol Exp (Wars) ; 81(4): 307-313, 2021.
Article in English | MEDLINE | ID: mdl-35014980

ABSTRACT

Store­operated calcium entry (SOCE) is important for refilling endoplasmic reticulum (ER) Ca2+ stores and Ca2+ signaling. Extracellular Ca2+ is conducted by highly Ca2+­selective Orai channels that are activated by stromal interaction molecule proteins (STIMs), which are sensitive ER Ca2+ sensors. We found an approximately five­fold increase in annexin A3a (anxa3a) expression levels in stim2b knockout zebrafish. The present study investigated whether annexin A3 protein is involved in SOCE. We used Ca2+ imaging and electrophysiological recordings to determine the effect of annexin A2, A3, and A6 overexpression on SOCE and Orai-dependent Ca2+ currents (ICRAC) in cultured cells. Annexin A3 increased SOCE amplitude and potentiated Ca2+ ICRAC currents. These results suggest that annexin A3 is a positive modulator of SOCE.


Subject(s)
Annexin A3/metabolism , Calcium Channels , Calcium , Animals , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Signaling , ORAI1 Protein , Stromal Interaction Molecule 1/genetics , Zebrafish/metabolism
14.
J Clin Lab Anal ; 35(2): e23622, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33118214

ABSTRACT

BACKGROUND: Melanoma, a relatively common malignancy, has become one of the tumors with the fastest rising incidence in recent years. The purpose of this study was to investigate the effect of Microglial Annexin A3 (ANXA3) on melanoma. METHODS: Serum samples were obtained from 20 patients with melanoma or 20 healthy controls. Kaplan-Meier survival analysis was performed. Transcriptome were used to analyze the correlation between ANXA3 expression and overall survival in patients with melanoma. Human melanoma cell lines WM-115 cells were transfected with ANXA3, si-ANXA3, ANXA3 + si-hypoxia inducible factor-1α (HIF-1α), si-ANXA3 + HIF-1α, and negative plasmids. Cell proliferation assay, cell invasion assay, and wound healing assay were performed on WM-115 cells. Lactate dehydrogenase (LDH) and caspase-3/9 activities were detected by commercial kits. Western blot and RT-PCR were used to detect the protein and mRNA expression of relation factors. RESULTS: ANXA3 expression was up-regulated in patients with melanoma in comparison with healthy controls. Over-expression of ANXA3 promoted cell growth and migration, and reduced cytotoxicity of WM-115 cells. Overall survival (OS) and disease-free survival (DFS) of patients with high ANXA3 expression were both lower than those of patients with low ANXA3 expression. Down-regulation of ANXA3 reduced cell growth and migration, and promoted cytotoxicity of WM-115 cells. ANXA3 induced vascular endothelial growth factor (VEGF) signaling pathway by activation of HIF-1α. CONCLUSION: In conclusion, our results indicated that ANXA3 promoted cell growth and migration of melanoma via activation of HIF-1α/VEGF signaling pathway.


Subject(s)
Annexin A3/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Melanoma/metabolism , Melanoma/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Vascular Endothelial Growth Factor A/metabolism , Annexin A3/blood , Annexin A3/genetics , Case-Control Studies , Cell Line, Tumor , Cell Movement , Humans , Melanoma/mortality , Skin Neoplasms/mortality , Melanoma, Cutaneous Malignant
15.
Int Immunopharmacol ; 89(Pt A): 107139, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33191179

ABSTRACT

Accumulating evidence has indicated that inflammation is required for the initiation and progression of hepatocellular carcinoma (HCC). The annexin family protein, which has a highly similar structure, has been demonstrated to participate in pro- or anti-inflammatory regulation in the developing of tumours. However, the potential effects of ANXA3 in the immune microenvironment of HCC remain unknown. In present study, we found that increased ANXA3 expression is associated with a higher infiltrated neutrophil-lymphocyte ratio (iNLR) in HCC. Moreover, HCC patients with a high iNLR and high ANXA3 expression confer the highest risk of death. ANXA3 can be detected in both cell lysates and culture supernatants. However, the secretory ANXA3 did not directly regulate the iNLR. Further study demonstrated that ANXA3 upregulated the iNLR by inducing chemokine CXCL8 and CCL25 release from HCC cells. We further confirmed that ANXA3 promotes tumourigenesis and detected the same associations between ANXA3 and the iNLR or chemokines in vivo. Our findings indicate that ANXA3 regulates the chemokine to remodel the iNLR and promotes tumourigenicity in HCC. These results further expanded our understanding of ANXA3 in the microenvironment of HCC and might provide novel targets for the investigation of molecular treatments for HCC patients.


Subject(s)
Annexin A3/metabolism , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Lymphocytes/physiology , Neutrophils/physiology , Tumor Microenvironment/immunology , Cell Line, Tumor , Cell Movement , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Up-Regulation/physiology
16.
Am J Pathol ; 190(11): 2317-2326, 2020 11.
Article in English | MEDLINE | ID: mdl-32861643

ABSTRACT

The adipocyte-like morphology of clear cell renal cell carcinoma (ccRCC) cells results from a grade-dependent neutral lipid accumulation; however, the molecular mechanism and role in renal cancer progression have yet to be clarified. ccRCC shows a gene expression signature consistent with adipogenesis, and the phospholipid-binding protein annexin A3 (AnxA3), a negative regulator of adipocyte differentiation, is down-regulated in RCC and shows a differential expression pattern for two isoforms of 36 and 33 kDa. Using primary cell cultures and cell lines, we investigated the involvement of AnxA3 isoforms in lipid storage modulation of ccRCC cells. We found that the increased accumulation of lipids into ccRCC cells correlated with a decrease of the 36/33 isoform ratio. Treatment with adipogenic medium induced a significant increment of lipid storage in ccRCC cells that had a low 36-kDa AnxA3 expression and 36/33 ratio. The 36-kDa AnxA3 silencing in ccRCC cells increased lipid storage induced by adipogenic medium. These data suggest that 36-kDa AnxA3 negatively modulates the response to adipogenic treatment and may act as negative regulator of lipid storage in ccRCC cells. The subcellular distribution of AnxA3 in the cellular endocytic compartment suggests its involvement in modulation of vesicular trafficking, and it might serve as a putative mechanism of lipid storage regulation in ccRCC cells, opening novel translational outcomes.


Subject(s)
Annexin A3/metabolism , Carcinoma, Renal Cell/metabolism , Kidney Neoplasms/metabolism , Lipid Metabolism , Neoplasm Proteins/metabolism , Aged , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Female , Humans , Isoenzymes/metabolism , Kidney Neoplasms/pathology , Male
17.
Immunology ; 161(4): 291-302, 2020 12.
Article in English | MEDLINE | ID: mdl-32682335

ABSTRACT

According to publicly available transcriptome datasets, the abundance of Annexin A3 (ANXA3) is robustly increased during the course of sepsis; however, no studies have examined the biological significance or clinical relevance of ANXA3 in this pathology. Here we explored this interpretation gap and identified possible directions for future research. Based on reference transcriptome datasets, we found that ANXA3 expression is restricted to neutrophils, is upregulated in vitro after exposure to plasma obtained from septic patients, and is associated with adverse clinical outcomes. Secondly, an increase in ANXA3 transcript abundance was also observed in vivo, in the blood of septic patients in multiple independent studies. ANXA3 is known to mediate calcium-dependent granules-phagosome fusion in support of microbicidal activity in neutrophils. More recent work has also shown that ANXA3 enhances proliferation and survival of tumour cells via a Caspase-3-dependent mechanism. And this same molecule is also known to play a critical role in regulation of apoptotic events in neutrophils. Thus, we posit that during sepsis ANXA3 might either play a beneficial role, by facilitating microbial clearance and resolution of the infection; or a detrimental role, by prolonging neutrophil survival, which is known to contribute to sepsis-mediated organ damage.


Subject(s)
Annexin A3/metabolism , Neutrophils/immunology , Sepsis/immunology , Access to Information , Animals , Annexin A3/genetics , Caspase 3/metabolism , Datasets as Topic , Humans , Phagosomes/metabolism , Transcriptome
18.
Eur Rev Med Pharmacol Sci ; 24(7): 3741-3748, 2020 04.
Article in English | MEDLINE | ID: mdl-32329851

ABSTRACT

OBJECTIVE: The purpose of this study was to explore the role of ANXA3 in lung cancer cell resistance to oxaliplatin (OXA). MATERIALS AND METHODS: After adding different concentrations of Ox, A549, and A549/Ox cell viability were examined using cell counting kit-8 (CCK-8) assay, and the mRNA and protein expressions of ANXA3 were analyzed by quantitative Real Time-Polymerase Chain Reaction (qRT-PCR) and Western blot, respectively. After treating cells with 5 µg/mL and 15 µg/mL Ox for 24 hours and knocking down ANXA3, qRT-PCR, CCK8, flow cytometry, transwell, and BrdU assays were performed to examine ANXA3 expression level, cell viability, apoptosis, migration, and proliferative capacities, respectively. In addition, Western blot was performed to detect the protein expression of c-caspase 3. RESULTS: The higher the concentration of Ox added, the worse the cell viability. Meanwhile, ANXA3 expression in A549/Ox cells was found remarkably higher than that in normal A549 cells. After treated with different concentrations of Ox for 24 hours, the cell viability, migration capacity and cell proliferation of A549 cells were found remarkably decreased, while the opposite results were observed in cell apoptosis and C-caspase 3 protein expression, and the Ox treatment group was evidently lower than control group. CONCLUSIONS: Knockdown of ANXA3 may be able to inhibit the resistance of LCa cells to OXA.


Subject(s)
Annexin A3/deficiency , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Oxaliplatin/pharmacology , A549 Cells , Annexin A3/genetics , Annexin A3/metabolism , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Lung Neoplasms/pathology , Tumor Cells, Cultured
19.
Dev Dyn ; 249(5): 666-678, 2020 05.
Article in English | MEDLINE | ID: mdl-32020697

ABSTRACT

BACKGROUND: Annexin A3 (Anxa3) is a member of the calcium-regulated, cell membrane-binding family of annexin proteins. We previously confirmed that Anxa3 is expressed in the endothelial lineage in vertebrates and that loss of anxa3 in Xenopus laevis leads to embryonic blood vessel defects. However, the biological function of Anxa3 in mammals is completely unknown. In order to investigate Anxa3 vascular function in mammals, we generated an endothelial cell-specific Anxa3 conditional knockout mouse model (Anxa3f/f ;Tie2-Cre). RESULTS: Anxa3f/f ;Tie2-Cre mice are born at Mendelian ratios and display morphologically normal blood vessels during development. However, loss of Anxa3 leads to artery-vein (AV) misalignment characterized by atypical AV crossovers in the postnatal and adult retina. CONCLUSIONS: Anxa3 is not essential for embryonic blood vessel formation but is required for proper parallel AV alignment in the murine retina. AV crossovers associated with Anxa3f/f ;Tie2-Cre mice are similar to AV intersections observed in patients with branch retinal vein occlusion (BRVO), although we did not observe occluded vessels. This new Anxa3 mouse model may provide a basis for understanding AV crossover formation associated with BRVO.


Subject(s)
Annexin A3/metabolism , Cell Movement/physiology , Cell Proliferation/physiology , Retina/metabolism , Veins/metabolism , Animals , Annexin A3/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Male , Mice , Retina/physiology , Veins/physiology
20.
FASEB J ; 34(4): 5144-5161, 2020 04.
Article in English | MEDLINE | ID: mdl-32043676

ABSTRACT

The development of the neuromuscular junction depends on signaling processes that involve protein phosphorylation. Motor neuron releases agrin to activate muscle protein Dok-7, a key tyrosine kinase essential for the formation of a mature and functional neuromuscular junction. However, the signaling cascade downstream of Dok-7 remains poorly understood. In this study, we combined the clustered regularly interspaced short palindromic repeats/Cas9 technique and quantitative phosphoproteomics analysis to study the tyrosine phosphorylation events triggered by agrin/Dok-7. We found tyrosine phosphorylation level of 36 proteins increased specifically by agrin stimulation. In Dok-7 mutant myotubes, however, 13 of the 36 proteins failed to be enhanced by agrin stimulation, suggesting that these 13 proteins are Dok-7-dependent tyrosine-phosphorylated proteins, could work as downstream molecules of agrin/Dok-7 signaling. We validated one of the proteins, Anxa3, by in vitro and in vivo assays. Knocking down of Anxa3 in the cultured myotubes inhibited agrin-induced AChR clustering, whereas reduction of Anxa3 in mouse muscles induced abnormal postsynaptic development. Collectively, our phosphoproteomics analysis provides novel insights into the complicated signaling network downstream of agrin/Dok-7.


Subject(s)
Agrin/physiology , Muscle Fibers, Skeletal/pathology , Muscle Proteins/physiology , Muscle, Skeletal/pathology , Neuromuscular Junction/pathology , Animals , Annexin A3/genetics , Annexin A3/metabolism , Mice , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Neuromuscular Junction/metabolism , Phosphoproteins , Phosphorylation , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL