1
|
Jiang M, Sun L, Jia Y, Ren X, Han L, Zhu Z, Zheng X. Causal effects of Annexin A1 and Annexin A2 on ischemic stroke and its subtypes: A two-sample Mendelian randomization study. J Cardiol 2025:S0914-5087(25)00099-1. [PMID: 40187529 DOI: 10.1016/j.jjcc.2025.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Preclinical studies have suggested that Annexin A1 and Annexin A2 act as anti-inflammatory agents, slowing the progression of atherosclerosis and further potentially reducing the risk of ischemic stroke. Since the causality of Annexins and ischemic stroke remains uncertain, this study aimed to investigate the causal effects of both using a two-sample Mendelian randomization (MR) method. METHODS The genetic instruments associated with Annexin A1 and Annexin A2 originated from a European-descent genome-wide association study (GWAS) of 50,000 participants from the INTERVAL study. Summary statistics for ischemic stroke and ischemic stroke subtypes were derived from the MEGASTROKE consortium's GWAS dataset, involving 40,585 cases and 406,111 controls of European ancestry. The inverse-variance weighted method was utilized in the main analysis, followed by a series of sensitivity analyses for robustness validation. RESULTS In the primary analysis, genetically predicted high Annexin A1 levels were associated with decreased risks of ischemic stroke (OR = 0.96; 95 % CI = 0.93-0.99; p = 0.023) and large artery stroke (OR = 0.88; 95 % CI = 0.81-0.96; p = 0.004). Similarly, genetically predicted high Annexin A2 levels also had significant associations with decreased risks of ischemic stroke (OR = 0.97; 95 % CI = 0.95-1.00; p = 0.019) and large artery stroke (OR = 0.90; 95 % CI = 0.85-0.96; p = 0.001). CONCLUSION In this two-sample MR study, we found that Annexins had causal protective effects against ischemic stroke, especially large artery stroke. Further basic mechanistic studies should be conducted to investigate the biological roles of these genes.
Collapse
Affiliation(s)
- Minglan Jiang
- Public Health Research Center and Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Lulu Sun
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Yiming Jia
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Xiao Ren
- Public Health Research Center and Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Longyang Han
- Public Health Research Center and Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Zhengbao Zhu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China.
| | - Xiaowei Zheng
- Public Health Research Center and Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China; Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
2
|
Sangamesh VC, Alagundagi DB, Jayaswamy PK, Kuriakose N, Shetty P. Targeting AnxA2-EGFR signaling: hydroxychloroquine as a therapeutic strategy for bleomycin-induced pulmonary fibrosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2015-2026. [PMID: 39222243 DOI: 10.1007/s00210-024-03417-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a disease that causes progressive failure of lung function, and its molecular mechanism remains poorly understood. However, the AnnexinA2-epidermal growth factor receptor (EGFR) signaling pathway has been identified as playing a significant role in its development. Hydroxychloroquine, a common anti-malarial drug, has been found to inhibit this pathway and slow down the progression of IPF. To better understand the role of the AnxA2-EGFR signaling pathway in pulmonary fibrosis, an in vivo study was conducted. In this study, mice were induced with pulmonary fibrosis using bleomycin, and HCQ was administered intraperitoneally the next day of bleomycin induction. The study also employed nintedanib as a positive control. After the induction, the lungs showed increased levels of fibronectin and vimentin, along with enhanced expression of AnxA2, EGFR, and Gal-3, indicating pulmonary fibrosis. Additionally, the study also found that HCQ significantly inhibited these effects and showed antifibrotic properties similar to nintedanib. Overall, these findings suggest that HCQ can attenuate bleomycin-induced pulmonary fibrosis by inhibiting the AnxA2-EGFR signaling pathway. These results are promising for developing new treatments for IPF.
Collapse
Affiliation(s)
- Vinay C Sangamesh
- Nitte University Centre for Science Education and Research, Nitte (Deemed to Be University), Deralakatte, Mangalore, 575018, Karnataka, India
| | - Dhananjay B Alagundagi
- Central Research Laboratory, KS. Hegde Medical Academy, Nitte (Deemed to Be University), Deralakatte, Mangalore, 575018, Karnataka, India
| | - Pavan K Jayaswamy
- Central Research Laboratory, KS. Hegde Medical Academy, Nitte (Deemed to Be University), Deralakatte, Mangalore, 575018, Karnataka, India
| | - Nithin Kuriakose
- Nitte University Centre for Science Education and Research, Nitte (Deemed to Be University), Deralakatte, Mangalore, 575018, Karnataka, India
| | - Praveenkumar Shetty
- Central Research Laboratory, KS. Hegde Medical Academy, Nitte (Deemed to Be University), Deralakatte, Mangalore, 575018, Karnataka, India.
- Department of Biochemistry, K.S. Hegde Medical Academy, Nitte (Deemed to Be University), Deralakatte, Mangalore, 575018, Karnataka, India.
| |
Collapse
|
3
|
Weijie S. Annexin A2: the feasibility of being a therapeutic target associated with cancer metastasis and drug resistance in cancer microenvironment. Discov Oncol 2024; 15:783. [PMID: 39692932 DOI: 10.1007/s12672-024-01693-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/10/2024] [Indexed: 12/19/2024] Open
Abstract
At present, there is still a lack of effective treatment strategies for cancer metastasis and drug resistance, so finding effective biomarkers is particularly important. AnnexinA2 (ANXA2), a vital membrane protein, critically influences cancer progression, tumor invasion, and tumor microenvironment modulation. To assess the possible application of ANXA2 as a therapeutic target against cancer cell metastasis and drug resistance to chemotherapeutic drugs in the tumor microenvironment, we elucidated the functionality of ANXA2 in stromal cells, angiogenic vascular cells, and infiltrated immune cells that mediate metastasis and drug resistance, as well as its potential as a therapeutic target. ANXA2 shows a high expression level in many tissues, and its expression level is even higher in several tumors and their microenvironments. ANXA2 is a crucial regulator of many factors and may serve as a target against drug-resistant cancers.
Collapse
Affiliation(s)
- Song Weijie
- Laboratory Animal Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Tianjin, 300060, China.
| |
Collapse
|
4
|
Fu R, Xu Y, Lu G, Zhang F, Liang P, Wang D. Identification and Immunological Characterization of Annexin B8 and Annexin E1 from Spirometra Erinaceieuropaei Spargana. Parasitol Res 2024; 123:398. [PMID: 39601902 DOI: 10.1007/s00436-024-08407-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024]
Abstract
Sparganosis is a parasitic zoonotic disease that poses a serious threat to public hygiene and human health. Annexin is a phospholipid-binding protein with calcium ion binding activity, serving various important functions, including interaction with the parasite and regulation of the host's immune response. In this study, two annexin (ANX) family genes, Spirometra erinaceieuropaei (S. erinaceieuropaei) Annexin B8 (SeANXB8) and E1 (SeANXE1), isolated from spargana, were cloned and immunologically characterized. Both recombinant S. erinaceieuropaei Annexin B8 (rSeANXB8) and E1 (rSeANXE1) were specifically recognized by serum from rats immunized with the recombinant proteins, displaying strong immunoreactivity. They are also among the major components of sparganum excretion/secretion products (ESPs). SeANXE1 was identified in the parasite's tegument, testis, genital pore, ovary, and eggs, while SeANXB8 was found in the parasite's tegument and eggs. Plasminogen (PLG)-binding assays revealed that the two annexins could bind to human PLG in a concentration-dependent manner, which was blocked by the corresponding antibodies. These findings suggest that SeANXB8 and SeANXE1 may be involved in host-parasite interaction and may influence the host's immune response during sparganosis. They could be potential diagnosis and vaccination targets for sparganosis.
Collapse
Affiliation(s)
- Ruijia Fu
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, Key Laboratory of Tropical Bioresources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, Hainan, China
- Key Laboratory of Tropical Translational Medicine of the Ministry of Education of China, Hainan Medical University, Haikou, 571199, Hainan, China
| | - Yanquan Xu
- Clinical Medicine Research Center, Xinqiao Hospital, Army Medical University, Chongqing, 40038, China
| | - Gang Lu
- Key Laboratory of Tropical Translational Medicine of the Ministry of Education of China, Hainan Medical University, Haikou, 571199, Hainan, China
| | - Fan Zhang
- Department of General Surgery, Hainan General Hospital, Hainan Medical University, Haikou, 570311, Hainan, China
| | - Pei Liang
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, Key Laboratory of Tropical Bioresources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, Hainan, China.
- Key Laboratory of Tropical Translational Medicine of the Ministry of Education of China, Hainan Medical University, Haikou, 571199, Hainan, China.
| | - Dayong Wang
- Laboratory of Biopharmaceuticals and Molecular Pharmacology, Key Laboratory of Tropical Bioresources of the Ministry of Education of China, School of Pharmaceutical Sciences, Hainan University, Haikou, 570228, Hainan, China.
- One Health Cooperative Innovation Center, Hainan University, Haikou, 570228, Hainan, China.
| |
Collapse
|
5
|
Christofidis K, Pergaris A, Fioretzaki R, Charalampakis N, Kapetanakis EΙ, Kavantzas N, Schizas D, Sakellariou S. Annexin A2 in Tumors of the Gastrointestinal Tract, Liver, and Pancreas. Cancers (Basel) 2024; 16:3764. [PMID: 39594718 PMCID: PMC11592865 DOI: 10.3390/cancers16223764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
Annexin A2 (ANXA2) is a protein that is involved in many physiological and pathological cellular processes. There is compelling evidence that its dysregulated expression, be it up- or downregulation, contributes to the oncogenesis of various neoplasms, including those of the digestive system. The present review summarizes the current knowledge on the role of ANXA2 in the main tumors of the digestive system. The clinical significance of ANXA2 is primordial, due to its potential use as a diagnostic and prognostic biomarker, and as a part of therapeutic protocols. Certain preclinical studies have shown that inhibiting ANXA2 or disrupting its interactions with key molecules can suppress tumor growth, invasion, and metastasis, as well as increase the cancer cells' sensitivity to treatment in various cancers. Further research is needed to fully elucidate the complex role of ANXA2 in the carcinogenesis of tumors of the digestive system, and to translate these findings into clinical applications for improved diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Konstantinos Christofidis
- Cytopathology Laboratory, Laiko General Hospital of Athens, 11527 Athens, Greece; (K.C.); (N.K.); (S.S.)
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Alexandros Pergaris
- First Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Rodanthi Fioretzaki
- First Department of Surgery, National and Kapodistrian University of Athens, Laiko General Hospital of Athens, 11527 Athens, Greece; (R.F.); (D.S.)
| | - Nikolaos Charalampakis
- Department of Medical Oncology, Metaxa Cancer Hospital of Piraeus, 18537 Piraeus, Greece;
| | - Emmanouil Ι. Kapetanakis
- Department of Thoracic Surgery, National and Kapodistrian University of Athens, Attikon University Hospital, 12462 Athens, Greece
| | - Nikolaos Kavantzas
- Cytopathology Laboratory, Laiko General Hospital of Athens, 11527 Athens, Greece; (K.C.); (N.K.); (S.S.)
| | - Dimitrios Schizas
- First Department of Surgery, National and Kapodistrian University of Athens, Laiko General Hospital of Athens, 11527 Athens, Greece; (R.F.); (D.S.)
| | - Stratigoula Sakellariou
- Cytopathology Laboratory, Laiko General Hospital of Athens, 11527 Athens, Greece; (K.C.); (N.K.); (S.S.)
| |
Collapse
|
6
|
White ZB, Nair S, Bredel M. The role of annexins in central nervous system development and disease. J Mol Med (Berl) 2024; 102:751-760. [PMID: 38639785 PMCID: PMC11106189 DOI: 10.1007/s00109-024-02443-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/20/2024]
Abstract
Annexins, a group of Ca2+-dependent phospholipid-binding proteins, exert diverse roles in neuronal development, normal central nervous system (CNS) functioning, neurological disorders, and CNS tumors. This paper reviews the roles of individual annexins (A1-A13) in these contexts. Annexins possess unique structural and functional features, such as Ca2+-dependent binding to phospholipids, participating in membrane organization, and modulating cell signaling. They are implicated in various CNS processes, including endocytosis, exocytosis, and stabilization of plasma membranes. Annexins exhibit dynamic roles in neuronal development, influencing differentiation, proliferation, and synaptic formation in CNS tissues. Notably, annexins such as ANXA1 and ANXA2 play roles in apoptosis and blood-brain barrier (BBB) integrity. Neurological disorders, including Alzheimer's disease, multiple sclerosis, and depression, involve annexin dysregulation, influencing neuroinflammation, blood-brain barrier integrity, and stress responses. Moreover, annexins contribute to the pathogenesis of CNS tumors, either promoting or suppressing tumor growth, angiogenesis, and invasion. Annexin expression patterns vary across different CNS tumor types, providing potential prognostic markers and therapeutic targets. This review underscores the multifaceted roles of annexins in the CNS, highlighting their importance in normal functioning, disease progression, and potential therapeutic interventions.
Collapse
Affiliation(s)
- Zachary B White
- Department of Radiation Oncology, O'Neal Comprehensive Cancer Center, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sindhu Nair
- Department of Radiation Oncology, O'Neal Comprehensive Cancer Center, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Markus Bredel
- Department of Radiation Oncology, O'Neal Comprehensive Cancer Center, Heersink School of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
7
|
Han G, Su Y, Mao Q, Han Z, Sun J. Identification and functional characterization of annexin A2 in half-smooth tongue sole (Cynoglossus semilaevis). FISH & SHELLFISH IMMUNOLOGY 2024; 148:109492. [PMID: 38467321 DOI: 10.1016/j.fsi.2024.109492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 03/13/2024]
Abstract
Annexin A2 (AnxA2), belonging to the annexin family, plays a crucial role in immune responses. In this study, the cDNA of the AnxA2 gene was identified in half-smooth tongue sole, Cynoglossus semilaevis. The transcript of AnxA2 gene in C. semilaevis (CsAnxA2) showed broad tissue distribution, with the highest expression level observed in the gut. CsAnxA2 expression was significantly up-regulated in the intestine, spleen, and kidney tissues following exposure to Shewanella algae. Immunohistochemical staining revealed that CsAnxA2 was predominantly expressed in epithelial cells and significantly elevated after S. algae challenge. Subcellular localization showed that CsAnxA2 was primarily localized in the cytoplasmic compartment. Moreover, proinflammatory cytokines (IL-6, IL-8 and IL-1β) exhibited significant upregulation after CsAnxA2 was overexpressed in vivo. One hundred and fifty-eight CsAnxA2-interacting proteins were captured in the intestinal tissue, showing the top two normalized abundance observed for actin beta (ACTB) and protein S100-A10 (p11). Fifty-four high abundance CsAnxA2-interacting proteins (HIPs) were primary enriched in ten pathways, with the top three significantly enriched pathways being Salmonella infection, glycolysis/gluconeogenesis, and peroxisome proliferator-activated receptor (PPAR) signaling pathway. These results provide valuable information for further investigation into the functional mechanism of AnxA2 in C. semilaevis.
Collapse
Affiliation(s)
- Guowei Han
- Key Laboratory of Smart Breeding (Co-construction by Ministry and Province), Tianjin Agricultural University, Tianjin, 300384, China; Tianjin Key Lab of Aqua-ecology and Aquaculture, Fisheries College, Tianjin Agricultural University, Tianjin, 300384, China.
| | - Yufeng Su
- Tianjin Key Lab of Aqua-ecology and Aquaculture, Fisheries College, Tianjin Agricultural University, Tianjin, 300384, China.
| | - Qing Mao
- Tianjin Key Lab of Aqua-ecology and Aquaculture, Fisheries College, Tianjin Agricultural University, Tianjin, 300384, China.
| | - Zhuoran Han
- Key Laboratory of Smart Breeding (Co-construction by Ministry and Province), Tianjin Agricultural University, Tianjin, 300384, China; Tianjin Key Lab of Aqua-ecology and Aquaculture, Fisheries College, Tianjin Agricultural University, Tianjin, 300384, China.
| | - Jingfeng Sun
- Key Laboratory of Smart Breeding (Co-construction by Ministry and Province), Tianjin Agricultural University, Tianjin, 300384, China; Tianjin Key Lab of Aqua-ecology and Aquaculture, Fisheries College, Tianjin Agricultural University, Tianjin, 300384, China.
| |
Collapse
|
8
|
Lei J, Sun P, Sheng J, Wang H, Xie Y, Song J. The intricate role of annexin A2 in kidney: a comprehensive review. Ren Fail 2023; 45:2273427. [PMID: 37955107 PMCID: PMC10653649 DOI: 10.1080/0886022x.2023.2273427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 10/16/2023] [Indexed: 11/14/2023] Open
Abstract
Annexin A2 (Anxa2) is a calcium (Ca2+)-regulated phospholipid binding protein composed of a variable N-terminus and a conserved core domain. This protein has been widely found in many tissues and fluids, including tubule cells, glomerular epithelial cells, renal vessels, and urine. In acute kidney injury, the expression level of this protein is markedly elevated in response to acute stress. Moreover, Anxa2 is a novel biomarker and potential therapeutic target with prognostic value in chronic kidney disease. In addition, Anxa2 is associated not only with clear-cell renal cell carcinoma differentiation but also the formation of calcium-related nephrolithiasis. In this review, we discuss the characteristics and functions of Anxa2 and focus on recent reports on the role of Anxa2 in the kidney, which may be useful for future research.
Collapse
Affiliation(s)
- Juan Lei
- Department of Pediatric Nephrology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Pingping Sun
- Department of Internal Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, P.R. China
| | - Jingyi Sheng
- Department of Pediatric Nephrology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Hongri Wang
- Department of Pediatric Nephrology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Yifan Xie
- Department of Rheumatism and Immunology, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| | - Jiayu Song
- Department of Pediatric Nephrology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, P.R. China
| |
Collapse
|
9
|
Siddiqui H, Deo N, Rutledge MT, Williams MJ, Redpath GM, McCormick SP. Plasminogen Receptors Promote Lipoprotein(a) Uptake by Enhancing Surface Binding and Facilitating Macropinocytosis. Arterioscler Thromb Vasc Biol 2023; 43:1851-1866. [PMID: 37589135 PMCID: PMC10521804 DOI: 10.1161/atvbaha.123.319344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 08/02/2023] [Indexed: 08/18/2023]
Abstract
BACKGROUND High levels of Lp(a) (lipoprotein(a)) are associated with multiple forms of cardiovascular disease. Lp(a) consists of an apoB100-containing particle attached to the plasminogen homologue apo(a). The pathways for Lp(a) clearance are not well understood. We previously discovered that the plasminogen receptor PlgRKT (plasminogen receptor with a C-terminal lysine) promoted Lp(a) uptake in liver cells. Here, we aimed to further define the role of PlgRKT and to investigate the role of 2 other plasminogen receptors, annexin A2 and S100A10 (S100 calcium-binding protein A10) in the endocytosis of Lp(a). METHODS Human hepatocellular carcinoma (HepG2) cells and haploid human fibroblast-like (HAP1) cells were used for overexpression and knockout of plasminogen receptors. The uptake of Lp(a), LDL (low-density lipoprotein), apo(a), and endocytic cargos was visualized and quantified by confocal microscopy and Western blotting. RESULTS The uptake of both Lp(a) and apo(a), but not LDL, was significantly increased in HepG2 and HAP1 cells overexpressing PlgRKT, annexin A2, or S100A10. Conversely, Lp(a) and apo(a), but not LDL, uptake was significantly reduced in HAP1 cells in which PlgRKT and S100A10 were knocked out. Surface binding studies in HepG2 cells showed that overexpression of PlgRKT, but not annexin A2 or S100A10, increased Lp(a) and apo(a) plasma membrane binding. Annexin A2 and S100A10, on the other hand, appeared to regulate macropinocytosis with both proteins significantly increasing the uptake of the macropinocytosis marker dextran when overexpressed in HepG2 and HAP1 cells and knockout of S100A10 significantly reducing dextran uptake. Bringing these observations together, we tested the effect of a PI3K (phosphoinositide-3-kinase) inhibitor, known to inhibit macropinocytosis, on Lp(a) uptake. Results showed a concentration-dependent reduction confirming that Lp(a) uptake was indeed mediated by macropinocytosis. CONCLUSIONS These findings uncover a novel pathway for Lp(a) endocytosis involving multiple plasminogen receptors that enhance surface binding and stimulate macropinocytosis of Lp(a). Although the findings were produced in cell culture models that have limitations, they could have clinical relevance since drugs that inhibit macropinocytosis are in clinical use, that is, the PI3K inhibitors for cancer therapy and some antidepressant compounds.
Collapse
Affiliation(s)
- Halima Siddiqui
- Department of Biochemistry (H.S., N.D., M.T.R., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
- School of Biomedical Sciences, HeartOtago (H.S., N.D., M.T.R., M.J.A.W., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
| | - Nikita Deo
- Department of Biochemistry (H.S., N.D., M.T.R., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
- School of Biomedical Sciences, HeartOtago (H.S., N.D., M.T.R., M.J.A.W., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
| | - Malcolm T. Rutledge
- Department of Biochemistry (H.S., N.D., M.T.R., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
- School of Biomedical Sciences, HeartOtago (H.S., N.D., M.T.R., M.J.A.W., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
| | - Michael J.A. Williams
- School of Biomedical Sciences, HeartOtago (H.S., N.D., M.T.R., M.J.A.W., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
- Department of Medicine (M.J.A.W.), Dunedin School of Medicine, University of Otago, New Zealand
| | - Gregory M.I. Redpath
- Department of Biochemistry (H.S., N.D., M.T.R., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
- School of Biomedical Sciences, HeartOtago (H.S., N.D., M.T.R., M.J.A.W., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
| | - Sally P.A. McCormick
- Department of Biochemistry (H.S., N.D., M.T.R., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
- School of Biomedical Sciences, HeartOtago (H.S., N.D., M.T.R., M.J.A.W., G.M.I.R., S.P.A.M.), Dunedin School of Medicine, University of Otago, New Zealand
| |
Collapse
|
10
|
Hosseini FA, Rejali L, Zabihi MR, Salehi Z, Daskar-Abkenar E, Taraz T, Fatemi N, Hashemi M, Asadzadeh-Aghdaei H, Nazemalhosseini-Mojarad E. Long non‑coding RNA LINC00460 contributes as a potential prognostic biomarker through its oncogenic role with ANXA2 in colorectal polyps. Mol Biol Rep 2023; 50:4505-4515. [PMID: 37024747 DOI: 10.1007/s11033-023-08393-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 03/17/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND Long intergenic non-coding RNA 460 (LINC00460) as a potential oncogene and Annexin A2 (ANXA2) as a promoter in different cancer progression processes was considered. A significant relationship between the LINC00460 and ANXA2 has been recently discovered in colorectal cancer (CRC). Therefore, defining molecular biomarkers accompanied by lesion histopathologic features can be a suggestive prognostic biomarker in precancerous polyps. This study aimed to investigate the elusive expression pattern of ANXA2 and LINC00460 in polyps. MATERIALS AND METHODS The construction of the co-expression and correlation network of LINC00460 and ANXA2 was plotted. LINC00460 and ANXA2 expression in 40 colon polyps was quantified by reverse transcription-real-time polymerase chain reaction. The receiver operating characteristic (ROC) curve was designed for distinguishing the high-risk precancerous lesion from the low-risk. Further, bioinformatics analysis was applied to find the shared MicroRNA-Interaction-Targets (MITs) between ANXA2 and LINC00460, and the associated pathways. RESULTS ANXA2 has a high co-expression rank with LINC00460 in the lncHUB database. Overexpression of ANXA2 and LINC00460 was distinguished in advanced adenoma polyps compared to the adjacent normal samples. The estimated AUC for ANXA2 and LINC00460 was 0.88 - 0.85 with 93%-90% sensitivity and 81%-70% specificity. In addition, eight MITs were shared between ANXA2 and LINC00460. Enrichment analysis detected several GO terms and pathways, including HIF-1α associated with cancer development. CONCLUSION In conclusion, the expression of the ANXA2 and LINC00460 were significantly elevated in pre-cancerous polyps, especially in high-risk adenomas. Collectively, ANXA2 and LINC00460 may be administered as potential prognostic biomarkers in patients with a precancerous large intestine lesion as an alarming issue.
Collapse
Affiliation(s)
- Farzaneh Alsadat Hosseini
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Leili Rejali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zabihi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi
- Laboratory of Complex Biological Systems and Bio-informatics (CBB), Department of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Elahe Daskar-Abkenar
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tannaz Taraz
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nayeralsadat Fatemi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence sciences Research Centre, Farhikhtegan Hospital, Islamic Azad University, Tehran Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh-Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ehsan Nazemalhosseini-Mojarad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Yeman St, Chamran Expressway, 19857-17413, Tehran, Iran.
| |
Collapse
|
11
|
Chowdhury SR, Koley T, Singh M, Samath EA, Kaur P. Association of Hsp90 with p53 and Fizzy related homolog (Fzr) synchronizing Anaphase Promoting Complex (APC/C): An unexplored ally towards oncogenic pathway. Biochim Biophys Acta Rev Cancer 2023; 1878:188883. [PMID: 36972769 DOI: 10.1016/j.bbcan.2023.188883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/29/2023]
Abstract
The intricate molecular interactions leading to the oncogenic pathway are the consequence of cell cycle modification controlled by a bunch of cell cycle regulatory proteins. The tumor suppressor and cell cycle regulatory proteins work in coordination to maintain a healthy cellular environment. The integrity of this cellular protein pool is perpetuated by heat shock proteins/chaperones, which assist in proper protein folding during normal and cellular stress conditions. Among these versatile groups of chaperone proteins, Hsp90 is one of the significant ATP-dependent chaperones that aid in stabilizing many tumor suppressors and cell cycle regulator protein targets. Recently, studies have revealed that in cancerous cell lines, Hsp90 stabilizes mutant p53, 'the guardian of the genome.' Hsp90 also has a significant impact on Fzr, an essential regulator of the cell cycle having an important role in the developmental process of various organisms, including Drosophila, yeast, Caenorhabditis elegans, and plants. During cell cycle progression, p53 and Fzr coordinately regulate the Anaphase Promoting Complex (APC/C) from metaphase to anaphase transition up to cell cycle exit. APC/C mediates proper centrosome function in the dividing cell. The centrosome acts as the microtubule organizing center for the correct segregation of the sister chromatids to ensure perfect cell division. This review examines the structure of Hsp90 and its co-chaperones, which work in synergy to stabilize proteins such as p53 and Fizzy-related homolog (Fzr) to synchronize the Anaphase Promoting Complex (APC/C). Dysfunction of this process activates the oncogenic pathway leading to the development of cancer. Additionally, an overview of current drugs targeting Hsp90 at various phases of clinical trials has been included.
Collapse
Affiliation(s)
- Sanghati Roy Chowdhury
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Tirthankar Koley
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Mandeep Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | | | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
12
|
Hsing DD, Stock AC, Greenwald BM, Bacha EA, Flynn PA, Carroll SJ, Dayton JD, Prockop SE, Qiu Y, Almeida D, Tamura S, Hajjar KA. Annexin A2 Loss After Cardiopulmonary Bypass and Development of Acute Postoperative Respiratory Dysfunction in Children. Crit Care Explor 2023; 5:e0862. [PMID: 36798534 PMCID: PMC9925105 DOI: 10.1097/cce.0000000000000862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
Abstract
The primary objective of this study was to determine whether expression of the multifunctional and adherens junction-regulating protein, annexin A2 (A2), is altered following cardiopulmonary bypass (CPB). A secondary objective was to determine whether depletion of A2 is associated with post-CPB organ dysfunction in children. DESIGN In a prospective, observational study conducted over a 1-year period in children undergoing cardiac surgery requiring CPB, we analyzed A2 expression in peripheral blood mononuclear cells at different time points. We then assessed the relationship of A2 expression with organ function at each time point in the early postoperative period. SETTING Twenty-three-bed mixed PICU in a tertiary academic center. PARTICIPANTS Patients 1 month to 18 years old undergoing cardiac surgery requiring CPB. MEAN OUTCOME MEASUREMENTS AND RESULTS We analyzed A2 expression in 22 enrolled subjects (n = 9, 1-23 mo old; n = 13, 2-18 yr old) and found a proteolysis-mediated decline in intact A2 immediately after bypass (p = 0.0009), reaching a median of 4% of baseline at 6 hours after bypass (p < 0.0001), and recovery by postoperative day 1. The degree of A2 depletion immediately after bypass in 1-23-month-olds correlated strongly with the extent of organ dysfunction, as measured by PICU admission Vasoactive-Ventilation-Renal (p = 0.004) and PEdiatric Logistic Organ Dysfunction-2 (p = 0.039) scores on postoperative day 1. A2 depletion immediately after bypass also correlated with more protracted requirement for both respiratory support (p = 0.007) and invasive ventilation (p = 0.013) in the 1-23-month-olds. CONCLUSIONS AND RELEVANCE The degree of depletion of A2 following CPB correlates with more severe organ dysfunction, especially acute respiratory compromise in children under 2 years. These findings suggest that loss of A2 may contribute to pulmonary microvascular leak in young children following CPB.
Collapse
Affiliation(s)
- Deyin D. Hsing
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Weill Cornell Medicine, New York City, NY
| | - Arabela C. Stock
- Division of Cardiac Critical Care Medicine, Heart Institute, Johns Hopkins All Children’s Hospital, St. Petersburg, FL
| | - Bruce M. Greenwald
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Weill Cornell Medicine, New York City, NY
| | - Emile A. Bacha
- Division of Cardiac, Thoracic and Vascular Surgery, Department of Surgery, Columbia University College of Physicians and Surgeons, New York City, NY
| | - Patrick A. Flynn
- Division of Pediatric Cardiology, Department of Pediatrics, Weill Cornell Medicine, New York City, NY
| | - Sheila J. Carroll
- Division of Pediatric Cardiology, Department of Pediatrics, Weill Cornell Medicine, New York City, NY
| | - Jeffrey D. Dayton
- Division of Pediatric Cardiology, Department of Pediatrics, Weill Cornell Medicine, New York City, NY
| | - Susan E. Prockop
- Stem Cell Transplant Program, Division of Hematology-Oncology, Boston Children’s Hospital, Department of Pediatric Oncology, Dana Farber Cancer Institute, Boston, MA
| | - Yuqing Qiu
- Division of Biostatistics and Epidemiology, Department of Population Health Sciences, Weill Cornell Medicine, New York City, NY
| | - Dena Almeida
- Division of Hematology-Oncology, Department of Pediatrics, Weill Cornell Medicine, New York City, NY
| | - Shoran Tamura
- Medical School, Class of 2024, Albert Einstein College of Medicine, Bronx, NY
| | - Katherine A. Hajjar
- Division of Hematology-Oncology, Department of Pediatrics, Weill Cornell Medicine, New York City, NY
| |
Collapse
|
13
|
Ning Y, Li Y, Wang H. ANXA2 is a potential biomarker for cancer prognosis and immune infiltration: A systematic pan-cancer analysis. Front Genet 2023; 14:1108167. [PMID: 36713082 PMCID: PMC9877333 DOI: 10.3389/fgene.2023.1108167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023] Open
Abstract
Background: Annexin A2 (ANXA2) belongs to the Annexin A family and plays a role in epithelial-mesenchymal transition, fibrinolysis, and other physiological processes. Annexin A2 has been extensively implicated in tumorigenesis and development in previous studies, but its precise role in pan-cancer remains largely unknown. Methods: We adopted bioinformatics methods to explore the oncogenic role of Annexin A2 using different databases, including the Cancer Genome Atlas (TCGA), the Genotype-Tissue Expression (GTEx) biobank, the Human Protein Atlas (HPA), the Gene Expression Profiling Interaction Analysis (GEPIA) and cBioPortal. We analyzed the differential expression of Annexin A2 in different tumors and its relationship with cancer prognosis, immune cell infiltration, DNA methylation, tumor mutation burden (TMB), microsatellite instability (MSI) and mismatch repair (MMR). Furtherly, we conducted a Gene Set Enrichment Analysis (GSEA) to identify the Annexin A2-related pathways. Results: Annexin A2 expression was upregulated in most cancers, except in kidney chromophobe (KICH) and prostate adenocarcinoma (PRAD). Annexin A2 showed a good diagnostic efficacy in twelve types of cancer. The high expression of Annexin A2 was significantly associated with a reduced overall survival, disease-specific survival and progression-free interval in seven cancers. The Annexin A2 expression was variably associated with infiltration of 24 types of immune cells in 32 tumor microenvironments. In addition, Annexin A2 expression was differently associated with 47 immune checkpoints, immunoregulators, DNA methylation, tumor mutation burden, microsatellite instability and mismatch repair in pan-cancer. Gene Set Enrichment Analysis revealed that Annexin A2 was significantly correlated with immune-related pathways in fifteen cancers. Conclusion: Annexin A2 widely correlates with immune infiltration and may function as a promising prognostic biomarker in many tumors, showing its potential as a target for immunotherapy in pan-cancer.
Collapse
Affiliation(s)
- Yijie Ning
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yufei Li
- Department of Neurosurgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Hongqin Wang
- Department of Neurosurgery, The First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
14
|
Porcù E, Maule F, Manfreda L, Mariotto E, Bresolin S, Cani A, Bortolozzi R, Della Puppa A, Corallo D, Viola G, Rampazzo E, Persano L. Identification of Homoharringtonine as a potent inhibitor of glioblastoma cell proliferation and migration. Transl Res 2023; 251:41-53. [PMID: 35788055 DOI: 10.1016/j.trsl.2022.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 11/18/2022]
Abstract
We previously demonstrated that Annexin A2 (ANXA2) is a pivotal mediator of the pro-oncogenic features displayed by glioblastoma (GBM) tumors, the deadliest adult brain malignancies, being involved in cell stemness, proliferation and invasion, thus negatively impacting patient prognosis. Based on these results, we hypothesized that compounds able to revert ANXA2-dependent transcriptional features could be exploited as reliable treatments to inhibit GBM cell aggressiveness by hampering their proliferative and migratory potential. Transcriptional signatures obtained by the modulation of ANXA2 activity/levels were functionally mapped through the QUADrATiC bioinformatic tool for compound identification. Selected compounds were screened by cell proliferation and migration assays in primary GBM cells, and we identified Homoharringtonine (HHT) as a potent inhibitor of GBM cell motility and proliferation, without affecting their viability. A further molecular characterization of the effects displayed by HHT, confirmed its ability to inhibit a transcriptional program involved in cell migration and invasion. Moreover, we demonstrated that the multiple antitumoral effects displayed by HHT are correlated to the inhibition of a platelet derived growth factor receptor α (PDGFRα)-dependent intracellular signaling through the impairment of Signal transducer and activator of transcription 3 (STAT3) and Ras homolog family member A (RhoA) axes. Our results demonstrate that HHT may act as a potent inhibitor of cancer cell proliferation and invasion in GBM, by hampering multiple PDGFRα-dependent oncogenic signals transduced through the STAT3 and RhoA intracellular components, finally suggesting its potential transferability for achieving an effective impairment of peculiar GBM hallmarks.
Collapse
Affiliation(s)
- Elena Porcù
- Department of Women and Children's Health, University of Padova, Padova, Italy; Pediatric Research Institute, Padova, Italy
| | - Francesca Maule
- Department of Women and Children's Health, University of Padova, Padova, Italy
| | - Lorenzo Manfreda
- Department of Women and Children's Health, University of Padova, Padova, Italy; Pediatric Research Institute, Padova, Italy
| | - Elena Mariotto
- Department of Women and Children's Health, University of Padova, Padova, Italy; Pediatric Research Institute, Padova, Italy
| | - Silvia Bresolin
- Department of Women and Children's Health, University of Padova, Padova, Italy; Pediatric Research Institute, Padova, Italy
| | - Alice Cani
- Department of Women and Children's Health, University of Padova, Padova, Italy; Pediatric Research Institute, Padova, Italy
| | | | | | - Diana Corallo
- Laboratory of Target Discovery and Biology of Neuroblastoma, Pediatric Research Institute, Padova, Italy
| | - Giampietro Viola
- Department of Women and Children's Health, University of Padova, Padova, Italy; Pediatric Research Institute, Padova, Italy
| | - Elena Rampazzo
- Department of Women and Children's Health, University of Padova, Padova, Italy; Pediatric Research Institute, Padova, Italy.
| | - Luca Persano
- Department of Women and Children's Health, University of Padova, Padova, Italy; Pediatric Research Institute, Padova, Italy
| |
Collapse
|
15
|
Encephalomyocarditis Virus 2A Protein Inhibited Apoptosis by Interaction with Annexin A2 through JNK/c-Jun Pathway. Viruses 2022; 14:v14020359. [PMID: 35215950 PMCID: PMC8880565 DOI: 10.3390/v14020359] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 01/28/2022] [Accepted: 02/07/2022] [Indexed: 11/16/2022] Open
Abstract
Encephalomyocarditis virus can cause myocarditis and encephalitis in pigs and other mammals, thus posing a potential threat to public health safety. The 2A protein is an important virulence factor of EMCV. Previous studies have shown that the 2A protein may be related to the inhibition of apoptosis by virus, but its specific molecular mechanism is not clear. In this study, the 2A protein was expressed in Escherichia coli in order to find interacting cell proteins. A pull down assay, coupled with mass spectrometry, revealed that the 2A protein possibly interacted with annexin A2. Co-immunoprecipitation assays and confocal imaging analysis further demonstrated that the 2A protein interacted with annexin A2 in cells. In reducing the expression of annexin A2 by siRNA, the ability of the 2A protein to inhibit apoptosis was weakened and the proliferation of EMCV was slowed down. These results suggest that annexin A2 is closely related to the inhibition of apoptosis by 2A. Furthermore, both RT-PCR and western blot results showed that the 2A protein requires annexin A2 interaction to inhibit apoptosis via JNK/c-Jun pathway. Taken together, our data indicate that the 2A protein inhibits apoptosis by interacting with annexin A2 via the JNK/c-Jun pathway. These findings provide insight into the molecular pathogenesis underlying EMCV infection.
Collapse
|
16
|
Mui L, Martin CM, Tschirhart BJ, Feng Q. Therapeutic Potential of Annexins in Sepsis and COVID-19. Front Pharmacol 2021; 12:735472. [PMID: 34566657 PMCID: PMC8458574 DOI: 10.3389/fphar.2021.735472] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/23/2021] [Indexed: 12/13/2022] Open
Abstract
Sepsis is a continuing problem in modern healthcare, with a relatively high prevalence, and a significant mortality rate worldwide. Currently, no specific anti-sepsis treatment exists despite decades of research on developing potential therapies. Annexins are molecules that show efficacy in preclinical models of sepsis but have not been investigated as a potential therapy in patients with sepsis. Human annexins play important roles in cell membrane dynamics, as well as mediation of systemic effects. Most notably, annexins are highly involved in anti-inflammatory processes, adaptive immunity, modulation of coagulation and fibrinolysis, as well as protective shielding of cells from phagocytosis. These discoveries led to the development of analogous peptides which mimic their physiological function, and investigation into the potential of using the annexins and their analogous peptides as therapeutic agents in conditions where inflammation and coagulation play a large role in the pathophysiology. In numerous studies, treatment with recombinant human annexins and annexin analogue peptides have consistently found positive outcomes in animal models of sepsis, myocardial infarction, and ischemia reperfusion injury. Annexins A1 and A5 improve organ function and reduce mortality in animal sepsis models, inhibit inflammatory processes, reduce inflammatory mediator release, and protect against ischemic injury. The mechanisms of action and demonstrated efficacy of annexins in animal models support development of annexins and their analogues for the treatment of sepsis. The effects of annexin A5 on inflammation and platelet activation may be particularly beneficial in disease caused by SARS-CoV-2 infection. Safety and efficacy of recombinant human annexin A5 are currently being studied in clinical trials in sepsis and severe COVID-19 patients.
Collapse
Affiliation(s)
- Louise Mui
- Division of Critical Care, Department of Medicine, Schulich School of Dentistry and Medicine, Western University, London, ON, Canada
| | - Claudio M Martin
- Division of Critical Care, Department of Medicine, Schulich School of Dentistry and Medicine, Western University, London, ON, Canada.,Lawson Health Research Institute, London Health Sciences Centre, London, ON, Canada
| | - Brent J Tschirhart
- Department of Physiology and Pharmacology, Schulich School of Dentistry and Medicine, Western University, London, ON, Canada
| | - Qingping Feng
- Lawson Health Research Institute, London Health Sciences Centre, London, ON, Canada.,Department of Physiology and Pharmacology, Schulich School of Dentistry and Medicine, Western University, London, ON, Canada
| |
Collapse
|
17
|
Birbo B, Madu EE, Madu CO, Jain A, Lu Y. Role of HSP90 in Cancer. Int J Mol Sci 2021; 22:ijms221910317. [PMID: 34638658 PMCID: PMC8508648 DOI: 10.3390/ijms221910317] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 11/25/2022] Open
Abstract
HSP90 is a vital chaperone protein conserved across all organisms. As a chaperone protein, it correctly folds client proteins. Structurally, this protein is a dimer with monomer subunits that consist of three main conserved domains known as the N-terminal domain, middle domain, and the C-terminal domain. Multiple isoforms of HSP90 exist, and these isoforms share high homology. These isoforms are present both within the cell and outside the cell. Isoforms HSP90α and HSP90β are present in the cytoplasm; TRAP1 is present in the mitochondria; and GRP94 is present in the endoplasmic reticulum and is likely secreted due to post-translational modifications (PTM). HSP90 is also secreted into an extracellular environment via an exosome pathway that differs from the classic secretion pathway. Various co-chaperones are necessary for HSP90 to function. Elevated levels of HSP90 have been observed in patients with cancer. Despite this observation, the possible role of HSP90 in cancer was overlooked because the chaperone was also present in extreme amounts in normal cells and was vital to normal cell function, as observed when the drastic adverse effects resulting from gene knockout inhibited the production of this protein. Differences between normal HSP90 and HSP90 of the tumor phenotype have been better understood and have aided in making the chaperone protein a target for cancer drugs. One difference is in the conformation: HSP90 of the tumor phenotype is more susceptible to inhibitors. Since overexpression of HSP90 is a factor in tumorigenesis, HSP90 inhibitors have been studied to combat the adverse effects of HSP90 overexpression. Monotherapies using HSP90 inhibitors have shown some success; however, combination therapies have shown better results and are thus being studied for a more effective cancer treatment.
Collapse
Affiliation(s)
- Bereket Birbo
- Massachusetts Institute of Technology, Cambridge, MA 02139, USA;
| | - Elechi E. Madu
- Departments of Biological Sciences, University of Memphis, Memphis, TN 38152, USA; (E.E.M.); (C.O.M.); (A.J.)
| | - Chikezie O. Madu
- Departments of Biological Sciences, University of Memphis, Memphis, TN 38152, USA; (E.E.M.); (C.O.M.); (A.J.)
| | - Aayush Jain
- Departments of Biological Sciences, University of Memphis, Memphis, TN 38152, USA; (E.E.M.); (C.O.M.); (A.J.)
| | - Yi Lu
- Health Science Center, Department of Pathology and Laboratory Medicine, University of Tennessee, Memphis, TN 38163, USA
- Correspondence: ; Tel.: +1-(901)-448-5436; Fax: +1-(901)-448-5496
| |
Collapse
|
18
|
Regimbeau M, Abrey J, Vautrot V, Causse S, Gobbo J, Garrido C. Heat shock proteins and exosomes in cancer theranostics. Semin Cancer Biol 2021; 86:46-57. [PMID: 34343652 DOI: 10.1016/j.semcancer.2021.07.014] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/21/2021] [Accepted: 07/21/2021] [Indexed: 01/19/2023]
Abstract
Heat shock proteins (HSPs) are a superfamily of molecular chaperones that were discovered through their ability to be induced by different stresses including heat shock. Other than their function as chaperones in proteins homeostasis, HSPs have been shown to inhibit different forms of cell death and to participate in cell proliferation and differentiation processes. Because cancer cells have to rewire their metabolism, they require a high amount of these stress-inducible chaperones for their survival. Therefore, HSPs are unusually abundant in cancer cells where they have oncogene-like functions. In cancer, HSPs have been involved in the regulation of apoptosis, immune responses, angiogenesis, metastasis and treatment resistance. Recently, HSPs have been shown to be secreted through exosomes by cancer cells. These tumor-derived exosomes can be used as circulating markers: HSP-exosomes have been reported as biomarkers of cancer dissemination, response to therapy and/or patient outcome. A new range of functions, mostly in modulation of anticancer immune responses, have been described for these extracellular HSPs. In this review, we will describe those recently reported functions of HSP-exosomes that makes them both targets for anticancer therapeutics and biomarkers for the monitoring of the disease. We will also discuss their emerging interest in cancer vaccines.
Collapse
Affiliation(s)
- Mathilde Regimbeau
- INSERM, UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC. 7 blvd Jeanne d'Arc, 21000, Dijon, France; Université. Bourgogne Franche-Comté, 21000, Dijon, France
| | - Jimena Abrey
- INSERM, UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC. 7 blvd Jeanne d'Arc, 21000, Dijon, France; Université. Bourgogne Franche-Comté, 21000, Dijon, France
| | - Valentin Vautrot
- INSERM, UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC. 7 blvd Jeanne d'Arc, 21000, Dijon, France; Université. Bourgogne Franche-Comté, 21000, Dijon, France; Anticancer Center Georges François Leclerc, Dijon, France
| | - Sebastien Causse
- INSERM, UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC. 7 blvd Jeanne d'Arc, 21000, Dijon, France; Université. Bourgogne Franche-Comté, 21000, Dijon, France
| | - Jessica Gobbo
- INSERM, UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC. 7 blvd Jeanne d'Arc, 21000, Dijon, France; Anticancer Center Georges François Leclerc, Dijon, France; Early Phase Unit INCa CLIP², Department of Oncology, Georges-François Leclerc Centre, Dijon, France; Centre d'investigation Clinique INSERM 1432, CHU Dijon-Bourgogne, Dijon, France
| | - Carmen Garrido
- INSERM, UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC. 7 blvd Jeanne d'Arc, 21000, Dijon, France; Université. Bourgogne Franche-Comté, 21000, Dijon, France; Anticancer Center Georges François Leclerc, Dijon, France.
| |
Collapse
|
19
|
Belvedere R, Morretta E, Pessolano E, Novizio N, Tosco A, Porta A, Whiteford J, Perretti M, Filippelli A, Monti MC, Petrella A. Mesoglycan exerts its fibrinolytic effect through the activation of annexin A2. J Cell Physiol 2021; 236:4926-4943. [PMID: 33284486 DOI: 10.1002/jcp.30207] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022]
Abstract
Mesoglycan is a drug based on a mixture of glycosaminoglycans mainly used for the treatment of blood vessel diseases acting as antithrombotic and profibrinolytic drugs. Besides the numerous clinical studies, there is no information about its function on the fibrinolytic cascade. Here, we have elucidated the mechanism of action by which mesoglycan induces the activation of plasmin from endothelial cells. Surprisingly, by a proteomic analysis, we found that, following mesoglycan treatment, these cells show a notable amount of annexin A2 (ANXA2) at the plasma membrane. This protein has been widely associated with fibrinolysis and appears able to move to the membrane when phosphorylated. In our model, this translocation has proven to enhance cell migration, invasion, and angiogenesis. Furthermore, the interaction of mesoglycan with syndecan 4 (SDC4), a coreceptor belonging to the class of heparan sulfate proteoglycans, represents the upstream event of the ANXA2 behavior. Indeed, the activation of SDC4 triggers the motility of endothelial cells culminating in angiogenesis. Interestingly, mesoglycan can induce the release of plasmin in endothelial cell supernatants only in the presence of ANXA2. This evaluation suggests that mesoglycan triggers the formation of a chain mechanism starting from the activation of SDC4, and the related cascade of events, including src complex and PKCα activation, promoting the phosphorylation of ANXA2 and its translocation to plasma membrane. This indicates a connection among mesoglycan, SDC4-(PKCα-src), and ANXA2 which, in turn, links the tissue plasminogen activator bringing it closer to plasminogen. This latter is so cleaved to release the plasmin and degrade fibrin sleeves.
Collapse
Affiliation(s)
| | - Elva Morretta
- Department of Pharmacy, University of Salerno, Fisciano (SA), Italy
| | - Emanuela Pessolano
- Department of Pharmacy, University of Salerno, Fisciano (SA), Italy
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nunzia Novizio
- Department of Pharmacy, University of Salerno, Fisciano (SA), Italy
| | - Alessandra Tosco
- Department of Pharmacy, University of Salerno, Fisciano (SA), Italy
| | - Amalia Porta
- Department of Pharmacy, University of Salerno, Fisciano (SA), Italy
| | - James Whiteford
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mauro Perretti
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Amelia Filippelli
- Department of Medicine, Surgery, and Dentistry, University of Salerno, Baronissi (SA), Italy
| | | | | |
Collapse
|
20
|
Cheng Y, Li W, Gui R, Wang C, Song J, Wang Z, Wang X, Shen Y, Wang Z, Hao L. Dual Characters of GH-IGF1 Signaling Pathways in Radiotherapy and Post-radiotherapy Repair of Cancers. Front Cell Dev Biol 2021; 9:671247. [PMID: 34178997 PMCID: PMC8220142 DOI: 10.3389/fcell.2021.671247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 05/17/2021] [Indexed: 12/02/2022] Open
Abstract
Radiotherapy remains one of the most important cancer treatment modalities. In the course of radiotherapy for tumor treatment, the incidental irradiation of adjacent tissues could not be completely avoided. DNA damage is one of the main factors of cell death caused by ionizing radiation, including single-strand (SSBs) and double-strand breaks (DSBs). The growth hormone-Insulin-like growth factor 1 (GH-IGF1) axis plays numerous roles in various systems by promoting cell proliferation and inhibiting apoptosis, supporting its effects in inducing the development of multiple cancers. Meanwhile, the GH-IGF1 signaling involved in DNA damage response (DDR) and DNA damage repair determines the radio-resistance of cancer cells subjected to radiotherapy and repair of adjacent tissues damaged by radiotherapy. In the present review, we firstly summarized the studies on GH-IGF1 signaling in the development of cancers. Then we discussed the adverse effect of GH-IGF1 signaling in radiotherapy to cancer cells and the favorable impact of GH-IGF1 signaling on radiation damage repair to adjacent tissues after irradiation. This review further summarized recent advances on research into the molecular mechanism of GH-IGF1 signaling pathway in these effects, expecting to specify the dual characters of GH-IGF1 signaling pathways in radiotherapy and post-radiotherapy repair of cancers, subsequently providing theoretical basis of their roles in increasing radiation sensitivity during cancer radiotherapy and repairing damage after radiotherapy.
Collapse
Affiliation(s)
- Yunyun Cheng
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Wanqiao Li
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Ruirui Gui
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Chunli Wang
- College of Animal Science, Jilin University, Changchun, China
| | - Jie Song
- College of Animal Science, Jilin University, Changchun, China
| | - Zhaoguo Wang
- College of Animal Science, Jilin University, Changchun, China
| | - Xue Wang
- The First Hospital of Jilin University, Changchun, China
| | - Yannan Shen
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Zhicheng Wang
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Linlin Hao
- College of Animal Science, Jilin University, Changchun, China
| |
Collapse
|
21
|
Li Z, Yu L, Hu B, Chen L, Jv M, Wang L, Zhou C, Wei M, Zhao L. Advances in cancer treatment: a new therapeutic target, Annexin A2. J Cancer 2021; 12:3587-3596. [PMID: 33995636 PMCID: PMC8120175 DOI: 10.7150/jca.55173] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/25/2021] [Indexed: 12/11/2022] Open
Abstract
Annexin A2 (ANXA2) is a calcium regulated phospholipid-binding protein. It is expressed in some tumor cells, endothelial cells, macrophages, and mononuclear cells, affecting cell survival and mediating interactions between intercellular and extracellular microenvironment. Aberrant expression of ANXA2 can be used as a potential predictive factor, diagnostic biomarker and therapeutic target in cancer therapy. Investigators used various technologies to target ANXA2 in a preclinical model of human cancers and demonstrated encouraging results. In this review article, we discuss the diagnosis and prognosis latent capacity of ANXA2 in progressive cancers, focus on the exploration of restorative interventions targeting ANXA2 in cancer treatment. Further, we comment on a promising candidate therapy that is conceivable for clinical translation.
Collapse
Affiliation(s)
- Zinan Li
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Lifeng Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Baohui Hu
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Lianze Chen
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Mingyi Jv
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Lin Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Chenyi Zhou
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Medical Diagnosis and Treatment Center, Liaoning Province, China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China.,Liaoning Engineering Technology Research Center, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang City, 110122, Liaoning, China
| |
Collapse
|
22
|
Fabbiano F, Corsi J, Gurrieri E, Trevisan C, Notarangelo M, D'Agostino VG. RNA packaging into extracellular vesicles: An orchestra of RNA-binding proteins? J Extracell Vesicles 2020; 10:e12043. [PMID: 33391635 PMCID: PMC7769857 DOI: 10.1002/jev2.12043] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 11/17/2020] [Accepted: 12/03/2020] [Indexed: 12/11/2022] Open
Abstract
Extracellular vesicles (EVs) are heterogeneous membranous particles released from the cells through different biogenetic and secretory mechanisms. We now conceive EVs as shuttles mediating cellular communication, carrying a variety of molecules resulting from intracellular homeostatic mechanisms. The RNA is a widely detected cargo and, impressively, a recognized functional intermediate that elects EVs as modulators of cancer cell phenotypes, determinants of disease spreading, cell surrogates in regenerative medicine, and a source for non-invasive molecular diagnostics. The mechanistic elucidation of the intracellular events responsible for the engagement of RNA into EVs will significantly improve the comprehension and possibly the prediction of EV "quality" in association with cell physiology. Interestingly, the application of multidisciplinary approaches, including biochemical as well as cell-based and computational strategies, is increasingly revealing an active RNA-packaging process implicating RNA-binding proteins (RBPs) in the sorting of coding and non-coding RNAs. In this review, we provide a comprehensive view of RBPs recently emerging as part of the EV biology, considering the scenarios where: (i) individual RBPs were detected in EVs along with their RNA substrates, (ii) RBPs were detected in EVs with inferred RNA targets, and (iii) EV-transcripts were found to harbour sequence motifs mirroring the activity of RBPs. Proteins so far identified are members of the hnRNP family (hnRNPA2B1, hnRNPC1, hnRNPG, hnRNPH1, hnRNPK, and hnRNPQ), as well as YBX1, HuR, AGO2, IGF2BP1, MEX3C, ANXA2, ALIX, NCL, FUS, TDP-43, MVP, LIN28, SRP9/14, QKI, and TERT. We describe the RBPs based on protein domain features, current knowledge on the association with human diseases, recognition of RNA consensus motifs, and the need to clarify the functional significance in different cellular contexts. We also summarize data on previously identified RBP inhibitor small molecules that could also be introduced in EV research as potential modulators of vesicular RNA sorting.
Collapse
Affiliation(s)
- Fabrizio Fabbiano
- Department of CellularComputational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Jessica Corsi
- Department of CellularComputational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Elena Gurrieri
- Department of CellularComputational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Caterina Trevisan
- Department of CellularComputational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Michela Notarangelo
- Department of CellularComputational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| | - Vito G. D'Agostino
- Department of CellularComputational and Integrative Biology (CIBIO)University of TrentoTrentoItaly
| |
Collapse
|
23
|
Expression of Annexin A2 Promotes Cancer Progression in Estrogen Receptor Negative Breast Cancers. Cells 2020; 9:cells9071582. [PMID: 32629869 PMCID: PMC7407301 DOI: 10.3390/cells9071582] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/22/2020] [Accepted: 06/27/2020] [Indexed: 12/26/2022] Open
Abstract
When breast cancer progresses to a metastatic stage, survival rates decline rapidly and it is considered incurable. Thus, deciphering the critical mechanisms of metastasis is of vital importance to develop new treatment options. We hypothesize that studying the proteins that are newly synthesized during the metastatic processes of migration and invasion will greatly enhance our understanding of breast cancer progression. We conducted a mass spectrometry screen following bioorthogonal noncanonical amino acid tagging to elucidate changes in the nascent proteome that occur during epidermal growth factor stimulation in migrating and invading cells. Annexin A2 was identified in this screen and subsequent examination of breast cancer cell lines revealed that Annexin A2 is specifically upregulated in estrogen receptor negative (ER-) cell lines. Furthermore, siRNA knockdown showed that Annexin A2 expression promotes the proliferation, wound healing and directional migration of breast cancer cells. In patients, Annexin A2 expression is increased in ER- breast cancer subtypes. Additionally, high Annexin A2 expression confers a higher probability of distant metastasis specifically for ER- patients. This work establishes a pivotal role of Annexin A2 in breast cancer progression and identifies Annexin A2 as a potential therapeutic target for the more aggressive and harder to treat ER- subtype.
Collapse
|
24
|
Beyene DA, Kanarek NF, Naab TJ, Ricks-Santi LL, Hudson TS. Annexin 2 protein expression is associated with breast cancer subtypes in African American women. Heliyon 2020; 6:e03241. [PMID: 32072035 PMCID: PMC7011040 DOI: 10.1016/j.heliyon.2020.e03241] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 09/18/2019] [Accepted: 01/14/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND A review of literature on the expression of Annexin 2 in cancer has shown that there is very limited research work on the association of this protein with breast cancer aggressiveness in African Americans. In the present study, TMA breast tissues from African American women were stained with Annexin 2 antibody to determine the association between the molecular subtypes and Annexin 2 protein expression. METHOD An annotated case series of 135 breast cancer tissues archived from 2000 to 2010 was acquired from the Howard University Tumor Registry. The association between ANX2 expression and survival by molecular subtypes Luminal A, Luminal B, HER2, and Triple Negative (TN) was assessed using Multinomial regression, chi-square analysis, and Kaplan-Meir graphs (Stata 11). RESULTS Our findings show a marked association between ANX2 protein expression in Luminal B and HER2 subtypes unadjusted and when adjusted for age. Borderline differences in tumor grade were found in TN only.Univariately, age (<50, 50 + years) and metastases were highly significant for overall survival, disease-free survival and recurrence-free survival. Stage, tumor size, and nodal involvement were of borderline or greater significance for overall and disease-free survival. ANX2 expression was not significant. Kaplan Meier tests of ANX2 showed significant separation of overall survival by ANX2 protein expression in all breast tumor subtypes. In multivariate analyses comparing TN to Luminal A, ANX2 was not important while controlling for age and grade. CONCLUSION ANX2 might be a biomarker of aggressiveness and a relevant candidate biomarker in high risk African American women with Luminal B and HER2 breast cancer.
Collapse
Affiliation(s)
- Desta A. Beyene
- Department of Biochemistry and Molecular Biology, Howard University, Washington, DC, USA
- Department of Research, Veteran Affairs Medical Center, Washington, DC, USA
- Howard University Cancer Center, Washington, DC, USA
| | - Norma F. Kanarek
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health and Department of Oncology, Johns Hospital School of Medicine, USA
| | - Tammey J. Naab
- Department of Pathology, College of Medicine, Howard University, Washington, DC, USA
| | - Luisel L. Ricks-Santi
- Department of Biological Sciences, Cancer Research Center, Hampton University, Hampton, VA, USA
| | - Tamaro S. Hudson
- Department of Research, Veteran Affairs Medical Center, Washington, DC, USA
- Department of Pharmacology, College of Medicine, Howard University, Washington, DC, USA
- Howard University Cancer Center, Washington, DC, USA
| |
Collapse
|
25
|
Annexin A2 interacting with ELMO1 regulates HCC chemotaxis and metastasis. Life Sci 2019; 222:168-174. [PMID: 30853625 DOI: 10.1016/j.lfs.2019.03.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/20/2019] [Accepted: 03/03/2019] [Indexed: 12/16/2022]
Abstract
AIMS SDF-1α induced chemotaxis plays an important role in hepatocellular carcinoma metastasis. CXCR4 stimulated by SDF-1α/CXCL12 triggers heterotrimeric G proteins activation, which regulate migration and chemotaxis of hepatocellular carcinoma cells. The pathways linking the chemokine GPCR/Gi signaling to actin polymerization for migration of cancer cells are not known. MATERIALS AND METHODS Through would healing assay, chemotaxis assay, F-actin polymerization assay, confocal assay, immunohistochemical assay, protein identification and coimmunoprecipitation assay, we detected the role and mechanisms of Annexin A2 in hepatocellular carcinoma. KEY FINDINGS In the present study, we firstly investigated the role of Annexin A2 in HepG2 cell chemotaxis and metastasis. Immunohistochemical analysis showed that Annexin A2 was highly expressed in hepatocellular carcinoma tissues. Its expression was closely associated with lymph node and distant metastasis. Knockdown Annexin A2 impaired cancer cell chemotaxis. Co-immunoprecipitation results showed an interaction between Annexin A2 and ELMO1. CXCL12 triggers an ELMO1-dependent membrane translocation of Annexin A2. SIGNIFICANCE Taken together, our results indicated an important role of Annexin A2 in hepatocellular carcinoma tissues metastasis and revealed a novel molecular mechanism of its activation.
Collapse
|
26
|
Aliyu IA, Ling KH, Md Hashim N, Chee HY. Annexin A2 extracellular translocation and virus interaction: A potential target for antivirus-drug discovery. Rev Med Virol 2019; 29:e2038. [PMID: 30746844 DOI: 10.1002/rmv.2038] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/20/2019] [Accepted: 01/21/2019] [Indexed: 12/23/2022]
Abstract
Annexin A2 is a membrane scaffolding and binding protein, which mediated various cellular events. Its functions are generally affected by cellular localization. In the cytoplasm, they interacted with different phospholipid membranes in Ca2+ -dependent manner and play vital roles including actin binding, remodeling and dynamics, cytoskeletal rearrangement, and lipid-raft microdomain formation. However, upon cell exposure to certain stimuli, annexin A2 translocates to the external leaflets of the plasma membrane where annexin A2 was recently reported to serve as a virus receptor, play an important role in the formation of virus replication complex, or implicated in virus assembly and budding. Here, we review some of annexin A2 roles in virus infections and the potentiality of targeting annexin A2 in the design of novel and promising antivirus agent that may have a broader consequence in virus therapy.
Collapse
Affiliation(s)
- Isah Abubakar Aliyu
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Science, University Putra Malaysia, Seri Kembangan, Malaysia.,Department of Medical Laboratory Science, Faculty of Allied Health Science, College of Health Science, Bayero University, Kano, Nigeria
| | - King-Hwa Ling
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, University Putra Malaysia, Seri Kembangan, Malaysia
| | - Nurfariesha Md Hashim
- Department of Biomedical Sciences, University Putra Malaysia, Seri Kembangan, Malaysia
| | - Hui-Yee Chee
- Department of Medical Microbiology and Parasitology, Faculty of Medicine and Health Science, University Putra Malaysia, Seri Kembangan, Malaysia
| |
Collapse
|
27
|
McCormick SPA, Schneider WJ. Lipoprotein(a) catabolism: a case of multiple receptors. Pathology 2018; 51:155-164. [PMID: 30595508 DOI: 10.1016/j.pathol.2018.11.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 10/31/2018] [Accepted: 11/01/2018] [Indexed: 01/09/2023]
Abstract
Lipoprotein(a) [Lp(a)] is an apolipoprotein B (apoB)-containing plasma lipoprotein similar in structure to low-density lipoprotein (LDL). Lp(a) is more complex than LDL due to the presence of apolipoprotein(a) [apo(a)], a large glycoprotein sharing extensive homology with plasminogen, which confers some unique properties onto Lp(a) particles. ApoB and apo(a) are essential for the assembly and catabolism of Lp(a); however, other proteins associated with the particle may modify its metabolism. Lp(a) specifically carries a cargo of oxidised phospholipids (OxPL) bound to apo(a) which stimulates many proinflammatory pathways in cells of the arterial wall, a key property underlying its pathogenicity and association with cardiovascular disease (CVD). While the liver and kidney are the major tissues implicated in Lp(a) clearance, the pathways for Lp(a) uptake appear to be complex and are still under investigation. Biochemical studies have revealed an exceptional array of receptors that associate with Lp(a) either via its apoB, apo(a), or OxPL components. These receptors fall into five main categories, namely 'classical' lipoprotein receptors, toll-like and scavenger receptors, lectins, and plasminogen receptors. The roles of these receptors have largely been dissected by genetic manipulation in cells or mice, although their relative physiological importance for removal of Lp(a) from the circulation remains unclear. The LPA gene encoding apo(a) has an overwhelming effect on Lp(a) levels which precludes any clear associations between potential Lp(a) receptor genes and Lp(a) levels in population studies. Targeted approaches and the selection of unique Lp(a) phenotypes within populations has nevertheless allowed for some associations to be made. Few of the proposed Lp(a) receptors can specifically be manipulated with current drugs and, as such, it is not currently clear whether any of these receptors could provide relevant targets for therapeutic manipulation of Lp(a) levels. This review summarises the current status of knowledge about receptor-mediated pathways for Lp(a) catabolism.
Collapse
Affiliation(s)
- Sally P A McCormick
- Department of Biochemistry, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.
| | - Wolfgang J Schneider
- Department of Medical Biochemistry, Max F. Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
28
|
S100A10 and Cancer Hallmarks: Structure, Functions, and its Emerging Role in Ovarian Cancer. Int J Mol Sci 2018; 19:ijms19124122. [PMID: 30572596 PMCID: PMC6321037 DOI: 10.3390/ijms19124122] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/04/2018] [Accepted: 12/17/2018] [Indexed: 12/25/2022] Open
Abstract
S100A10, which is also known as p11, is located in the plasma membrane and forms a heterotetramer with annexin A2. The heterotetramer, comprising of two subunits of annexin A2 and S100A10, activates the plasminogen activation pathway, which is involved in cellular repair of normal tissues. Increased expression of annexin A2 and S100A10 in cancer cells leads to increased levels of plasmin—which promotes the degradation of the extracellular matrix—increased angiogenesis, and the invasion of the surrounding organs. Although many studies have investigated the functional role of annexin A2 in cancer cells, including ovarian cancer, S100A10 has been less studied. We recently demonstrated that high stromal annexin A2 and high cytoplasmic S100A10 expression is associated with a 3.4-fold increased risk of progression and 7.9-fold risk of death in ovarian cancer patients. Other studies have linked S100A10 with multidrug resistance in ovarian cancer; however, no functional studies to date have been performed in ovarian cancer cells. This article reviews the current understanding of S100A10 function in cancer with a particular focus on ovarian cancer.
Collapse
|
29
|
Yang J, Kong P, Yang J, Jia Z, Hu X, Wang Z, Cui H, Bi Y, Qian Y, Li H, Wang F, Yang B, Yan T, Ma Y, Zhang L, Cheng C, Song B, Li Y, Xu E, Liu H, Gao W, Wang J, Liu Y, Zhai Y, Chang L, Wang Y, Zhang Y, Shi R, Liu J, Wang Q, Cheng X, Cui Y. High TSTA3 Expression as a Candidate Biomarker for Poor Prognosis of Patients With ESCC. Technol Cancer Res Treat 2018; 17:1533033818781405. [PMID: 29950151 PMCID: PMC6048620 DOI: 10.1177/1533033818781405] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Esophageal squamous cell carcinoma is the sixth most lethal cancer worldwide and the
fourth most lethal cancer in China. Tissue-specific transplantation antigen P35B codifies
the enzyme GDP-d-mannose-4,6-dehydratase, which participates in the biosynthesis
of GDP-l-fucose. GDP-l-fucose is an important substrate involved in the
biosynthesis of many glycoproteins. Cancer cells are often accompanied by the changes in
glycoprotein structure, which affects the adhesion, invasion, and metastasis of cells. It
is not clear whether tissue-specific transplantation antigen P35B has any effect on the
development of esophageal squamous cell carcinoma. We used an immunohistochemical method
to assess the expression of tissue-specific transplantation antigen P35B in 104 esophageal
squamous cell carcinoma samples. The results showed tissue-specific transplantation
antigen P35B expression was associated with some clinical features in patients, such as
age (P = .017), clinical stage (P = .010), and lymph
node metastasis (P = .043). Kaplan-Meier analysis and log-rank test
showed that patients with esophageal squamous cell carcinoma having high tissue-specific
transplantation antigen P35B expression had a worse prognosis compared to the patients
with low expression (P = .048). Multivariate Cox proportional hazards
regression model showed that high expression of tissue-specific transplantation antigen
P35B could predict poor prognosis for patients with esophageal squamous cell carcinoma
independently. In conclusion, abnormal fucosylation might participate in the progress of
esophageal squamous cell carcinoma and tissue-specific transplantation antigen P35B may
serve as a novel biomarker for prognosis of patients with esophageal squamous cell
carcinoma.
Collapse
Affiliation(s)
- Jie Yang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,3 Department of Gastroenterology, The Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Pengzhou Kong
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Jian Yang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Zhiwu Jia
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Xiaoling Hu
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,4 Department of Pharmacology, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Zianyi Wang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,5 Taiyuan Lingde Secondary School, Taiyuan, Shanxi, PR China
| | - Heyang Cui
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yanghui Bi
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yu Qian
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Hongyi Li
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Fang Wang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Bin Yang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,6 Department of General Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi, PR China
| | - Ting Yan
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yanchun Ma
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Ling Zhang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Caixia Cheng
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,7 Department of Pathology, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Bin Song
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,8 Department of Oncology, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yaoping Li
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,9 Department of Anorectum, Shanxi Provincial People's Hospital, Taiyuan, Shanxi, China
| | - Enwei Xu
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,10 Department of Pathology, Shanxi Cancer Hospital, Taiyuan, Shanxi, PR China
| | - Haiyan Liu
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Wei Gao
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,11 Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, Taiyuan, Shanxi, PR China.,12 Department of Otolaryngology Head & Neck Surgery, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Juan Wang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yiqian Liu
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yuanfang Zhai
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,13 Department of Anatomy, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Lu Chang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yi Wang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yingchun Zhang
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,13 Department of Anatomy, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Ruyi Shi
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Jing Liu
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,14 Department of General Surgery, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Qi Wang
- 3 Department of Gastroenterology, The Second Hospital, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Xiaolong Cheng
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China.,13 Department of Anatomy, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yongping Cui
- 1 Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, PR China.,2 Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| |
Collapse
|
30
|
Sharma MC. Annexin A2 (ANX A2): An emerging biomarker and potential therapeutic target for aggressive cancers. Int J Cancer 2018; 144:2074-2081. [PMID: 30125343 DOI: 10.1002/ijc.31817] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 08/08/2018] [Accepted: 08/13/2018] [Indexed: 12/12/2022]
Abstract
ANX A2 is an important member of annexin family of proteins expressed on surface of endothelial cells (ECs), macrophages, mononuclear cells and various types of cancer cells. It exhibits high affinity binding for calcium (Ca++ ) and phospholipids. ANX A2 plays an important role in many biological processes such as endocytosis, exocytosis, autophagy, cell-cell communications and biochemical activation of plasminogen. On the cell surface ANX A2 organizes the assembly of plasminogen (PLG) and tissue plasminogen activator (tPA) for efficient conversion of PLG to plasmin, a serine protease. Proteolytic activity of plasmin is required for activation of inactive pro-metalloproteases (pro-MMPs) and latent growth factors for their biological actions. These activation steps are critical for degradation of extracellular matrix (ECM) and basement proteins (BM) for cancer cell invasion and metastasis. Increased expression of ANX A2 protein/gene has been correlated with invasion and metastasis in a variety of human cancers. Moreover, clinical studies have positively correlated ANX A2 protein expression with aggressive cancers and with resistance to anticancer drugs, shorter disease-free survival (DFS), and worse overall survival (OS). The mechanism(s) by which ANX A2 regulates cancer invasion and metastasis are beginning to emerge. Investigators used various technologies to target ANX A2 in preclinical model of human cancers and demonstrated exciting results. In this review article, we analyzed existing literature concurrent with our own findings and provided a critical overview of ANX A2-dependent mechanism(s) of cancer invasion and metastasis.
Collapse
Affiliation(s)
- Mahesh C Sharma
- Research Service, Veterans Affairs Medical Center, Washington, DC.,Department of Biochemistry and Molecular Medicine, George Washington University, Washington, DC
| |
Collapse
|
31
|
Su H, Yang X, Wang S, Shi H, Liu X. Effect of annexin II-mediated conversion of plasmin from plasminogen on airborne transmission of H9N2 avian influenza virus. Vet Microbiol 2018; 223:100-106. [PMID: 30173734 DOI: 10.1016/j.vetmic.2018.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 08/01/2018] [Accepted: 08/02/2018] [Indexed: 11/16/2022]
Abstract
Airborne transmission plays an important role in dissemination of H9N2 subtype avian influenza virus. Annexin II (A2)-mediated activation of plasminogen (PLG) promotes cleavage of the influenza virus HA protein and viral replication, resulting in enhanced pathogenesis. In this study, airborne transmission competent and defective strains of H9N2 influenza virus, SH7 and SH14, respectively, were used to investigate the effect of A2 on airborne spread. The results showed that A2 protein was increased in SH7 virions compared with SH14 particles, the binding ability of the SH7-infected MDCK cells to PLG was significantly higher than the SH14-infected cells, and influence efficiency of the PLG on replicated ability of SH7 virus was significantly stronger than that of SH14 virus, who spread without airborne route, indicating that the annexin 2 (A2) can bind PLG and contributes to SH7 with high replication ability. Furthermore, the copies of SH7 in the airborne infected chickens under inhibited by 6-AHA were significantly decreased, suggesting that the release of H9N2 avian influenza virus were reduced by inhibiting the conversion of PLG to PL, ultimately resulting in reduced airborne transmission of H9N2 avian influenza virus. In summary, A2-mediated conversion of PLG to PL plays a role in the airborne transmission capacity of H9N2 avian influenza viruses, and this interaction may represent potential targets for prevention and treatment of influenza virus infection.
Collapse
Affiliation(s)
- Hailong Su
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| | - Xueqin Yang
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| | - Shifeng Wang
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611-0880, USA.
| | - Huoying Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| | - Xiufan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, Jiangsu, China; Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China.
| |
Collapse
|
32
|
Shergalis A, Bankhead A, Luesakul U, Muangsin N, Neamati N. Current Challenges and Opportunities in Treating Glioblastoma. Pharmacol Rev 2018; 70:412-445. [PMID: 29669750 PMCID: PMC5907910 DOI: 10.1124/pr.117.014944] [Citation(s) in RCA: 560] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM), the most common and aggressive primary brain tumor, has a high mortality rate despite extensive efforts to develop new treatments. GBM exhibits both intra- and intertumor heterogeneity, lending to resistance and eventual tumor recurrence. Large-scale genomic and proteomic analysis of GBM tumors has uncovered potential drug targets. Effective and "druggable" targets must be validated to embark on a robust medicinal chemistry campaign culminating in the discovery of clinical candidates. Here, we review recent developments in GBM drug discovery and delivery. To identify GBM drug targets, we performed extensive bioinformatics analysis using data from The Cancer Genome Atlas project. We discovered 20 genes, BOC, CLEC4GP1, ELOVL6, EREG, ESR2, FDCSP, FURIN, FUT8-AS1, GZMB, IRX3, LITAF, NDEL1, NKX3-1, PODNL1, PTPRN, QSOX1, SEMA4F, TH, VEGFC, and C20orf166AS1 that are overexpressed in a subpopulation of GBM patients and correlate with poor survival outcomes. Importantly, nine of these genes exhibit higher expression in GBM versus low-grade glioma and may be involved in disease progression. In this review, we discuss these proteins in the context of GBM disease progression. We also conducted computational multi-parameter optimization to assess the blood-brain barrier (BBB) permeability of small molecules in clinical trials for GBM treatment. Drug delivery in the context of GBM is particularly challenging because the BBB hinders small molecule transport. Therefore, we discuss novel drug delivery methods, including nanoparticles and prodrugs. Given the aggressive nature of GBM and the complexity of targeting the central nervous system, effective treatment options are a major unmet medical need. Identification and validation of biomarkers and drug targets associated with GBM disease progression present an exciting opportunity to improve treatment of this devastating disease.
Collapse
Affiliation(s)
- Andrea Shergalis
- Department of Medicinal Chemistry, College of Pharmacy, North Campus Research Complex, Ann Arbor, Michigan (A.S., U.L., N.N.); Biostatistics Department and School of Public Health, University of Michigan, Ann Arbor, Michigan (A.B.); and Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand (U.L., N.M.)
| | - Armand Bankhead
- Department of Medicinal Chemistry, College of Pharmacy, North Campus Research Complex, Ann Arbor, Michigan (A.S., U.L., N.N.); Biostatistics Department and School of Public Health, University of Michigan, Ann Arbor, Michigan (A.B.); and Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand (U.L., N.M.)
| | - Urarika Luesakul
- Department of Medicinal Chemistry, College of Pharmacy, North Campus Research Complex, Ann Arbor, Michigan (A.S., U.L., N.N.); Biostatistics Department and School of Public Health, University of Michigan, Ann Arbor, Michigan (A.B.); and Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand (U.L., N.M.)
| | - Nongnuj Muangsin
- Department of Medicinal Chemistry, College of Pharmacy, North Campus Research Complex, Ann Arbor, Michigan (A.S., U.L., N.N.); Biostatistics Department and School of Public Health, University of Michigan, Ann Arbor, Michigan (A.B.); and Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand (U.L., N.M.)
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, North Campus Research Complex, Ann Arbor, Michigan (A.S., U.L., N.N.); Biostatistics Department and School of Public Health, University of Michigan, Ann Arbor, Michigan (A.B.); and Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand (U.L., N.M.)
| |
Collapse
|
33
|
Jiménez-Altayó F, Meirelles T, Crosas-Molist E, Sorolla MA, Del Blanco DG, López-Luque J, Mas-Stachurska A, Siegert AM, Bonorino F, Barberà L, García C, Condom E, Sitges M, Rodríguez-Pascual F, Laurindo F, Schröder K, Ros J, Fabregat I, Egea G. Redox stress in Marfan syndrome: Dissecting the role of the NADPH oxidase NOX4 in aortic aneurysm. Free Radic Biol Med 2018; 118:44-58. [PMID: 29471108 DOI: 10.1016/j.freeradbiomed.2018.02.023] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/12/2018] [Accepted: 02/15/2018] [Indexed: 11/16/2022]
Abstract
Marfan syndrome (MFS) is characterized by the formation of ascending aortic aneurysms resulting from altered assembly of extracellular matrix fibrillin-containing microfibrils and dysfunction of TGF-β signaling. Here we identify the molecular targets of redox stress in aortic aneurysms from MFS patients, and investigate the role of NOX4, whose expression is strongly induced by TGF-β, in aneurysm formation and progression in a murine model of MFS. Working models included aortae and cultured vascular smooth muscle cells (VSMC) from MFS patients, and a NOX4-deficient Marfan mouse model (Fbn1C1039G/+-Nox4-/-). Increased tyrosine nitration and reactive oxygen species levels were found in the tunica media of human aortic aneurysms and in cultured VSMC. Proteomic analysis identified nitrated and carbonylated proteins, which included smooth muscle α-actin (αSMA) and annexin A2. NOX4 immunostaining increased in the tunica media of human Marfan aorta and was transcriptionally overexpressed in VSMC. Fbn1C1039G/+-Nox4-/- mice aortas showed a reduction of fragmented elastic fibers, which was accompanied by an amelioration in the Marfan-associated enlargement of the aortic root. Increase in the contractile phenotype marker calponin in the tunica media of MFS mice aortas was abrogated in Fbn1C1039G/+-Nox4-/- mice. Endothelial dysfunction evaluated by myography in the Marfan ascending aorta was prevented by the absence of Nox4 or catalase-induced H2O2 decomposition. We conclude that redox stress occurs in MFS, whose targets are actin-based cytoskeleton members and regulators of extracellular matrix homeostasis. Likewise, NOX4 have an impact in the progression of the aortic dilation in MFS and in the structural organization of the aortic tunica media, the VSMC phenotypic modulation, and endothelial function.
Collapse
Affiliation(s)
- Francesc Jiménez-Altayó
- Departament de Farmacologia, Terapèutica i Toxicologia, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Thayna Meirelles
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Eva Crosas-Molist
- TGF-β and Cancer Group, Oncobell Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - M Alba Sorolla
- Institut de Recerca Biomèdica de Lleida (IRB Lleida), Lleida, Spain; Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, Lleida, Spain
| | - Darya Gorbenko Del Blanco
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Judit López-Luque
- TGF-β and Cancer Group, Oncobell Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Ana-Maria Siegert
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Fabio Bonorino
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Laura Barberà
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain
| | - Carolina García
- Department of Pathology, Hospital de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Barcelona, and Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
| | - Enric Condom
- Department of Pathology, Hospital de Bellvitge-IDIBELL, L'Hospitalet de Llobregat, Barcelona, and Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain
| | - Marta Sitges
- Cardiovascular Institute, Hospital Clinic, IDIBAPS-University of Barcelona, Barcelona, Spain
| | | | - Francisco Laurindo
- Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - Katrin Schröder
- German Center of Cardiovascular Research (DZHK), Partner site Rhein Main, Frankfurt am Main, Germany
| | - Joaquim Ros
- Institut de Recerca Biomèdica de Lleida (IRB Lleida), Lleida, Spain; Departament de Ciències Mèdiques Bàsiques, Universitat de Lleida, Lleida, Spain
| | - Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain; Department de Ciències Fisiològiques, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Gustavo Egea
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, and IDIBAPS, Barcelona, Spain.
| |
Collapse
|
34
|
Annexin 2A sustains glioblastoma cell dissemination and proliferation. Oncotarget 2018; 7:54632-54649. [PMID: 27429043 PMCID: PMC5342369 DOI: 10.18632/oncotarget.10565] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 06/03/2016] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma (GBM) is the most devastating tumor of the brain, characterized by an almost inevitable tendency to recur after intensive treatments and a fatal prognosis. Indeed, despite recent technical improvements in GBM surgery, the complete eradication of cancer cell disseminated outside the tumor mass still remains a crucial issue for glioma patients management. In this context, Annexin 2A (ANXA2) is a phospholipid-binding protein expressed in a variety of cell types, whose expression has been recently associated with cell dissemination and metastasis in many cancer types, thus making ANXA2 an attractive putative regulator of cell invasion also in GBM. Here we show that ANXA2 is over-expressed in GBM and positively correlates with tumor aggressiveness and patient survival. In particular, we associate the expression of ANXA2 to a mesenchymal and metastatic phenotype of GBM tumors. Moreover, we functionally characterized the effects exerted by ANXA2 inhibition in primary GBM cultures, demonstrating its ability to sustain cell migration, matrix invasion, cytoskeletal remodeling and proliferation. Finally, we were able to generate an ANXA2-dependent gene signature with a significant prognostic potential in different cohorts of solid tumor patients, including GBM. In conclusion, we demonstrate that ANXA2 acts at multiple levels in determining the disseminating and aggressive behaviour of GBM cells, thus proving its potential as a possible target and strong prognostic factor in the future management of GBM patients.
Collapse
|
35
|
Yang W, Mei FC, Cheng X. EPAC1 regulates endothelial annexin A2 cell surface translocation and plasminogen activation. FASEB J 2018; 32:2212-2222. [PMID: 29217666 DOI: 10.1096/fj.201701027r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Annexins, a family of highly conserved calcium- and phospholipid-binding proteins, play important roles in a wide range of physiologic functions. Among the 12 known annexins in humans, annexin A2 (AnxA2) is one of the most extensively studied and has been implicated in various human diseases. AnxA2 can exist as a monomer or a heterotetrameric complex with S100A10 (P11) and plays a critical role in many cellular processes, including exocytosis, endocytosis, and membrane organization. At the endothelial cell surface, the (AnxA2⋅P11)2 tetramer-acting as a coreceptor for plasminogen and tissue plasminogen activator (tPA)-accelerates tPA-dependent activation of the fibrinolytic protease, plasmin, the enzyme that is responsible for thrombus dissolution and the degradation of fibrin. This study demonstrates that EPAC1 (exchange proteins directly activated by cAMP isoform 1) interacts with AnxA2 and regulates its biologic functions by modulating its membrane translocation in endothelial cells. By using genetic and pharmacologic approaches, we demonstrate that EPAC1-acting via the PLCε-PKC pathway-inhibits AnxA2 surface translocation and plasminogen activation. These results suggest that EPAC1 plays a role in the regulation of fibrinolysis in endothelial cells and may represent a novel therapeutic target for disorders of fibrinolysis.-Yang, W., Mei, F. C., Cheng, X. EPAC1 regulates endothelial annexin A2 cell surface translocation and plasminogen activation.
Collapse
Affiliation(s)
- Wenli Yang
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, USA
| | - Fang C Mei
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, USA
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Texas Therapeutics Institute, University of Texas Health Science Center, Houston, Texas, USA
| |
Collapse
|
36
|
Stromal Annexin A2 expression is predictive of decreased survival in pancreatic cancer. Oncotarget 2017; 8:106405-106414. [PMID: 29290958 PMCID: PMC5739743 DOI: 10.18632/oncotarget.22433] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 10/25/2017] [Indexed: 01/18/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) is renowned for high rates of metastasis and poor survival. Its notoriously dense fibrotic stroma contributes to chemoresistance. Stromal signaling in PDA is recognized for its multiple roles in regulating tumor invasion and metastasis. However, no stromal biomarker which can predict survival in PDA exists. Annexin A2 (AnxA2) was formerly identified as a metastasis-associated protein in PDA and tumoral overexpression is associated with poor survival. In this study, we examined AnxA2 expression in the tumor microenvironment in a preclinical model of PDA which suggests its role in tumor colonization. We injected wild-type (KPC) and AnxA2 knockout (KPCA) pancreatic cells into C57BL/GJ (B6) and AnxA2 knockout (KO) mice using the hemi-spleen model and observed their survival. We performed quantitative immunohistochemistry examining stromal AnxA2 expression in 56 patients who had surgically resected PDA and correlated expression with clinical outcomes. B6 mice injected with KPC cells demonstrated decreased median survival compared to those injected with KPCA cells (90 days vs. not reached, p < 0.0001) whereas there was no survival difference in the AnxA2 KO mice (p = 0.63). In patient specimens, we found that high stromal AnxA2 expression (≥80th percentile) was associated with significantly reduced disease-free survival (p = 0.002) and overall survival (p < 0.001). Using multivariate Cox models, there were no significant associations between other clinical covariates apart from high stromal AnxA2 expression. This study highlights the role that stromal AnxA2 expression plays as a prognostic marker in PDA and its potential as a predictive biomarker for survival outcomes in PDA.
Collapse
|
37
|
Zhao S, Su G, Yang W, Yue P, Bai B, Lin Y, Zhang J, Ba Y, Luo Z, Liu X, Zhao L, Xie Y, Xu Y, Li S, Meng W, Xie X, Li X. Identification and Comparison of Differentiation-Related Proteins in Hepatocellular Carcinoma Tissues by Proteomics. Technol Cancer Res Treat 2017; 16:1092-1101. [PMID: 29332450 PMCID: PMC5762076 DOI: 10.1177/1533034617732426] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Histological differentiation is a major pathological criterion indicating the risk of tumor invasion and metastasis in patients with hepatocellular carcinoma. The degree of tumor differentiation is controlled by a complex interacting network of associated proteins. The principal aim of the present study is to identify the possible differentiation-related proteins which may be used for early diagnosis and more effective therapies. We compared poorly differentiated and well-differentiated hepatocellular carcinoma tissues by using 2-dimensional gel electrophoresis and matrix-assisted laser desorption ionization time-of-flight mass spectrometry. Among the 11 identified protein spots, 6 were found to be upregulated in poorly differentiated hepatocellular carcinoma tissues and 5 were correspondingly downregulated. Immunohistochemistry was performed on 106 hepatocellular carcinoma tissues to confirm the results of the proteomic analysis. By using bioinformatic tools GO and STRING, these proteins were found to be related to catalytic activity, binding, and antioxidant activity. In particular, our data suggest that overexpression of peroxiredoxin-2, annexin A2, and heat shock protein β-1 was correlated with tumor invasion, metastasis, and poor prognosis, and therefore, these proteins may serve as potential diagnostic and therapeutic biomarkers.
Collapse
Affiliation(s)
- Sheng Zhao
- 1 Special Minimally Invasive Surgery, The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,2 Hepatopancreatobiliary Surgery Institute of Gansu Province, Clinical Medical College Cancer Center of Lanzhou University, Lanzhou, Gansu, China.,3 Biological Therapy and Regenerative Medicine Transformation Center of Gansu Province, Lanzhou, Gansu, China.,4 Department of General Surgery, Petrochemical General Hospital of Lanzhou, Lanzhou, Gansu, China
| | - Gang Su
- 5 Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Wenke Yang
- 5 Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Ping Yue
- 1 Special Minimally Invasive Surgery, The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,2 Hepatopancreatobiliary Surgery Institute of Gansu Province, Clinical Medical College Cancer Center of Lanzhou University, Lanzhou, Gansu, China.,3 Biological Therapy and Regenerative Medicine Transformation Center of Gansu Province, Lanzhou, Gansu, China
| | - Bing Bai
- 1 Special Minimally Invasive Surgery, The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,2 Hepatopancreatobiliary Surgery Institute of Gansu Province, Clinical Medical College Cancer Center of Lanzhou University, Lanzhou, Gansu, China.,3 Biological Therapy and Regenerative Medicine Transformation Center of Gansu Province, Lanzhou, Gansu, China
| | - Yanyan Lin
- 1 Special Minimally Invasive Surgery, The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,2 Hepatopancreatobiliary Surgery Institute of Gansu Province, Clinical Medical College Cancer Center of Lanzhou University, Lanzhou, Gansu, China.,3 Biological Therapy and Regenerative Medicine Transformation Center of Gansu Province, Lanzhou, Gansu, China
| | - Jinduo Zhang
- 1 Special Minimally Invasive Surgery, The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,2 Hepatopancreatobiliary Surgery Institute of Gansu Province, Clinical Medical College Cancer Center of Lanzhou University, Lanzhou, Gansu, China.,3 Biological Therapy and Regenerative Medicine Transformation Center of Gansu Province, Lanzhou, Gansu, China
| | - Yongjiang Ba
- 1 Special Minimally Invasive Surgery, The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,2 Hepatopancreatobiliary Surgery Institute of Gansu Province, Clinical Medical College Cancer Center of Lanzhou University, Lanzhou, Gansu, China.,3 Biological Therapy and Regenerative Medicine Transformation Center of Gansu Province, Lanzhou, Gansu, China
| | - Zhiwen Luo
- 1 Special Minimally Invasive Surgery, The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,2 Hepatopancreatobiliary Surgery Institute of Gansu Province, Clinical Medical College Cancer Center of Lanzhou University, Lanzhou, Gansu, China.,3 Biological Therapy and Regenerative Medicine Transformation Center of Gansu Province, Lanzhou, Gansu, China
| | - Xiaoming Liu
- 4 Department of General Surgery, Petrochemical General Hospital of Lanzhou, Lanzhou, Gansu, China
| | - Lili Zhao
- 6 The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yi Xie
- 6 The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yaowei Xu
- 6 The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Shuo Li
- 1 Special Minimally Invasive Surgery, The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Wenbo Meng
- 1 Special Minimally Invasive Surgery, The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,2 Hepatopancreatobiliary Surgery Institute of Gansu Province, Clinical Medical College Cancer Center of Lanzhou University, Lanzhou, Gansu, China.,3 Biological Therapy and Regenerative Medicine Transformation Center of Gansu Province, Lanzhou, Gansu, China
| | - Xiaodong Xie
- 5 Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou, Gansu, China
| | - Xun Li
- 1 Special Minimally Invasive Surgery, The First Clinical Medical College of Lanzhou University, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.,2 Hepatopancreatobiliary Surgery Institute of Gansu Province, Clinical Medical College Cancer Center of Lanzhou University, Lanzhou, Gansu, China.,3 Biological Therapy and Regenerative Medicine Transformation Center of Gansu Province, Lanzhou, Gansu, China.,7 The Second Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
38
|
Chen YD, Fang YT, Cheng YL, Lin CF, Hsu LJ, Wang SY, Anderson R, Chang CP, Lin YS. Exophagy of annexin A2 via RAB11, RAB8A and RAB27A in IFN-γ-stimulated lung epithelial cells. Sci Rep 2017; 7:5676. [PMID: 28720835 PMCID: PMC5516008 DOI: 10.1038/s41598-017-06076-4] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 06/07/2017] [Indexed: 12/09/2022] Open
Abstract
Annexin A2 (ANXA2), a phospholipid-binding protein, has multiple biological functions depending on its cellular localization. We previously demonstrated that IFN-γ-triggered ANXA2 secretion is associated with exosomal release. Here, we show that IFN-γ-induced autophagy is essential for the extracellular secretion of ANXA2 in lung epithelial cells. We observed colocalization of ANXA2-containing autophagosomes with multivesicular bodies (MVBs) after IFN-γ stimulation, followed by exosomal release. IFN-γ-induced exophagic release of ANXA2 could not be observed in ATG5-silenced or mutant RAB11-expressing cells. Furthermore, knockdown of RAB8A and RAB27A, but not RAB27B, reduced IFN-γ-triggered ANXA2 secretion. Surface translocation of ANXA2 enhanced efferocytosis by epithelial cells, and inhibition of different exophagic steps, including autophagosome formation, fusion of autophagosomes with MVBs, and fusion of amphisomes with plasma membrane, reduced ANXA2-mediated efferocytosis. Our data reveal a novel route of IFN-γ-induced exophagy of ANXA2.
Collapse
Affiliation(s)
- Ying-Da Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ting Fang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Lin Cheng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chiou-Feng Lin
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Li-Jin Hsu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shu-Ying Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Robert Anderson
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Departments of Microbiology & Immunology and Pediatrics, and Canadian Center for Vaccinology, Dalhousie University, Halifax, Canada
| | - Chih-Peng Chang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Yee-Shin Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
39
|
Ilich A, Bokarev I, Key NS. Global assays of fibrinolysis. Int J Lab Hematol 2017; 39:441-447. [PMID: 28497494 DOI: 10.1111/ijlh.12688] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 03/22/2017] [Indexed: 12/22/2022]
Abstract
Fibrinolysis is an important and integral part of the hemostatic system. Acting as a balance to blood coagulation, the fibrinolytic system protects the body from unwanted thrombus formation and occlusion of blood vessels. As long as blood coagulation and fibrinolysis remain in equilibrium, response to injury, such as vessel damage, is appropriately regulated. However, alterations in this balance may lead to thrombosis or bleeding. A variety of methods have been proposed to assess fibrinolytic activity in blood or its components, but due to the complexity of the system, the design of a "gold standard" assay that reflects overall fibrinolysis has remained an elusive goal. In this review, we describe the most commonly used methods that have been described, such as thromboelastography (TEG and ROTEM), global fibrinolytic capacity in plasma and whole blood, plasma turbidity methods, simultaneous thrombin and plasmin generation assays, euglobulin clot lysis time and fibrin plate methods. All of these assays have strengths and limitations. We suggest that some methods may be preferable for detecting hypofibrinolytic conditions, whereas others may be better for detecting hyperfibrinolytic states.
Collapse
Affiliation(s)
- A Ilich
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Divisions of Internal Medicine 1, Department of Medicine, First Moscow State Medical University n.a. I.M.Sechenov, Moscow, Russia
| | - I Bokarev
- Divisions of Cardiology, Department of Medicine, First Moscow State Medical University n.a. I.M.Sechenov, Moscow, Russia
| | - N S Key
- Division of Hematology/Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
40
|
Li DH, He CR, Liu FP, Li J, Gao JW, Li Y, Xu WD. Annexin A2, up-regulated by IL-6, promotes the ossification of ligament fibroblasts from ankylosing spondylitis patients. Biomed Pharmacother 2016; 84:674-679. [DOI: 10.1016/j.biopha.2016.09.091] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 09/22/2016] [Accepted: 09/22/2016] [Indexed: 12/17/2022] Open
|
41
|
Salle V, Schmidt J, Smail A, Mazière C, Conte MA, Brulé A, Mazière JC, Cadet E, Herpe YE, Duhaut P. Antibodies directed against annexin A2 and obstetric morbidity. J Reprod Immunol 2016; 118:50-53. [PMID: 27631133 DOI: 10.1016/j.jri.2016.08.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 08/25/2016] [Accepted: 08/29/2016] [Indexed: 01/11/2023]
Abstract
Acquired and inherited thrombophilia have both been reported to be associated with an increased risk of obstetric complications in early or later stages of pregnancy. Annexin A2 (ANXA2) is strongly expressed in vascular and placental tissues and plays a crucial role in fibrinolysis. The aim of the present study was to evaluate the prevalence of antibodies directed against ANXA2 in patients with recurrent miscarriage or obstetric complications. Anti-ANXA2 antibodies (aANXA2) were detected by ELISA in the sera from 46 women with obstetric morbidity, mainly recurrent miscarriage. The cut-off value for positivity was defined as 3 standard deviations above the mean optical density (OD) obtained in the sera from 42 female blood donors. The prevalence of aANXA2 in patients and healthy individuals was 15.2% and 2.3%, respectively. A statistically significant difference was observed between the 2 groups in terms of aANXA2 IgG titers (p=0.01). The highest aANXA2 levels were observed in sera from 2 patients with recurrent miscarriage and one patient with preeclampsia. aANXA2 could play a role in thrombotic mechanisms leading to recurrent pregnancy loss and placental vascular disease. Further studies are needed to determine whether ANXA2 is critical for maintenance of placental integrity.
Collapse
Affiliation(s)
- V Salle
- Department of Internal Medicine, Amiens University Hospital, France; INSERM U1088, Biochemistry Laboratory, Amiens University Hospital, France.
| | - J Schmidt
- Department of Internal Medicine, Amiens University Hospital, France
| | - A Smail
- Department of Internal Medicine, Amiens University Hospital, France
| | - C Mazière
- INSERM U1088, Biochemistry Laboratory, Amiens University Hospital, France
| | - M A Conte
- INSERM U1088, Biochemistry Laboratory, Amiens University Hospital, France
| | - A Brulé
- French Blood Establishment-North of France, France
| | - J C Mazière
- INSERM U1088, Biochemistry Laboratory, Amiens University Hospital, France
| | - E Cadet
- Department of Genetics, Amiens University Hospital, France
| | - Y E Herpe
- Biobank of Picardie, Amiens University Hospital, Amiens, France
| | - P Duhaut
- Department of Internal Medicine, Amiens University Hospital, France
| |
Collapse
|
42
|
Wang T, Yuan J, Zhang J, Tian R, Ji W, Zhou Y, Yang Y, Song W, Zhang F, Niu R. Anxa2 binds to STAT3 and promotes epithelial to mesenchymal transition in breast cancer cells. Oncotarget 2016; 6:30975-92. [PMID: 26307676 PMCID: PMC4741582 DOI: 10.18632/oncotarget.5199] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Accepted: 08/09/2015] [Indexed: 12/31/2022] Open
Abstract
Overexpression of annexin A2 (Anxa2) is correlated with invasion and metastasis in breast cancer cells. In this study, breast cancer patients with upregulated Anxa2 exhibited poor overall and disease-free survival rates. Anxa2 expression was also positively correlated with the expression of epidermal growth factor receptor (EGFR) and epithelial–mesenchymal transition (EMT) markers in breast cancer tissues and cell lines. Moreover, knockdown of Anxa2 impaired EGF-induced EMT, as well as the migration and invasion of breast cancer cells in vitro. Meanwhile, Anxa2 depletion significantly ablated pulmonary metastasis in a severe combined immunodeficiency mouse model of breast cancer. Importantly, Anxa2 reduction inhibited EGF-induced activation of STAT3, which is required for EGF-induced EMT. Anxa2 directly bound to STAT3 and enhanced its transcriptional activity, thereby indicating that Anxa2 promotes EGF-induced EMT in a STAT3-dependent manner. Our findings provide clinical evidence that Anxa2 is a poor prognostic factor for breast cancer and reveal a novel mechanism through which Anxa2 promotes breast cancer metastasis.
Collapse
Affiliation(s)
- Tong Wang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, The Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Jie Yuan
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, The Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Jie Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, The Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Ran Tian
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, The Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Wei Ji
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, The Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Yan Zhou
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, The Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Yi Yang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, The Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Weijie Song
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, The Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Fei Zhang
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, The Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| | - Ruifang Niu
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, The Key Laboratory of Breast Cancer Prevention and Therapy, Ministry of Education, Key Laboratory of Cancer Prevention and Therapy, Tianjin, PR China
| |
Collapse
|
43
|
Annexin A2 Coordinates STAT3 to Regulate the Invasion and Migration of Colorectal Cancer Cells In Vitro. Gastroenterol Res Pract 2016; 2016:3521453. [PMID: 27274723 PMCID: PMC4870365 DOI: 10.1155/2016/3521453] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 02/11/2016] [Accepted: 03/24/2016] [Indexed: 01/01/2023] Open
Abstract
The present study aimed to reveal the expression of STAT3 and Anxa 2 in CRC specimens and to investigate the effects of STAT3 and Anxa 2 signaling on the proliferation, invasion, and migration in CRC Caco-2 cells. Results demonstrated that both Anxa 2 and STAT3 were highly expressed in CRC specimens in both mRNA and protein levels, with or without phosphorylation (Tyrosine 23 in Anxa 2 and Tyrosine 705 in STAT3). And the upregulated Anxa 2 promoted the phosphorylation of STAT3 (Tyrosine 705) in CRC Caco-2 cells. The upregulated Anxa 2 promoted the proliferation, migration, and invasion of Caco-2 cells in vitro. Moreover, the STAT3 knockdown also repressed the proliferation, migration, and invasion of Caco-2 cells. In conclusion, the overexpressed Annexin A2 regulated the proliferation, invasion, and migration in CRC cells in an association with STAT3.
Collapse
|
44
|
Sharma MC, Tuszynski GP, Blackman MR, Sharma M. Long-term efficacy and downstream mechanism of anti-annexinA2 monoclonal antibody (anti-ANX A2 mAb) in a pre-clinical model of aggressive human breast cancer. Cancer Lett 2016; 373:27-35. [PMID: 26797420 DOI: 10.1016/j.canlet.2016.01.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 01/06/2016] [Accepted: 01/07/2016] [Indexed: 01/09/2023]
Abstract
There is considerable direct evidence that calcium binding protein ANX A2 is a potential target for treating aggressive breast cancer. The most compelling data are based on the finding of ANX A2 overexpression in aggressive triple negative human breast cancer (TNBC) cell lines and in human breast cancer tissues. Previously, we and others reported a unique role of ANX A2 in cancer invasion, including breast cancer. Moreover, we demonstrated that anti-ANX A2 mAb-mediated immunoneutralization of ANX A2 inhibited invasive human breast cancer growth in a xenograft model. We further evaluated the long-term effects of multiple treatments with anti-ANX A2 mAb and its mechanism of inhibition on human breast tumor growth. We now demonstrate that three treatments with anti-ANX A2 mAb led to significant inhibition of breast tumor growth in immunodeficient mice, and that the anti-tumor response was demonstrable from day 94. After treatment, we followed tumor growth for 172 days and demonstrated 67% inhibition of tumor growth without detectable adverse effects. Biochemical analysis demonstrated that anti-ANX A2 mAb treatment caused significant inhibition of conversion of tissue plasminogen activator (tPA) in the tumor microenvironment. This led to disruption of plasmin generation that consequently inhibited activation of MMP-9 and MMP-2. These results suggest that ANX A2 plays an important role in aggressive breast tumor growth by regulating proteolytic pathways in the tumor microenvironment. ANX A2 may represent a new target for the development of therapeutics for treatment of aggressive breast cancer.
Collapse
Affiliation(s)
- Mahesh C Sharma
- Research Service, Veterans Affairs Medical Center, Washington, DC 20422, USA; Department of Biochemistry and Molecular Medicine, George Washington University, Washington, DC, USA.
| | - George P Tuszynski
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Marc R Blackman
- Research Service, Veterans Affairs Medical Center, Washington, DC 20422, USA; Department of Biochemistry and Molecular Medicine, George Washington University, Washington, DC, USA; Department of Medicine, George Washington University, Washington, DC, USA
| | - Meena Sharma
- University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
45
|
Salle V, Cordonnier C, Schmidt J, Mazière C, Smail A, Attencourt C, Mabille MP, Mazière JC, Makdassi R, Choukroun G, Diouf M, Duhaut P, Ducroix JP. Vascular expression of annexin A2 in lupus nephritis. J Clin Pathol 2015; 69:533-6. [PMID: 26511441 DOI: 10.1136/jclinpath-2015-203139] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 10/05/2015] [Indexed: 11/04/2022]
Abstract
AIMS To evaluate vascular expression of annexin A2 (ANXA2) and its subunit S100A10 in lupus nephritis (LN). METHODS The present histological study included 14 patients with LN and 11 controls (patients with non-lupus kidney diseases). Kidney biopsies from patients with lupus were scored for lupus glomerulonephritis (according to the International Society of Nephrology/Renal Pathology Society 2003 classification) and vascular lesions (such as microthrombi and antiphospholipid syndrome nephropathy (APSN)). ANXA2 and S100A10 expression in glomerular and peritubular capillaries was evaluated by immunohistochemistry on tissue sections. The staining intensity score ranged from 0 (no expression) to 4 (intense expression). RESULTS In patients with LN, the median age (range) at first kidney biopsy was 36 (18-49). Vascular lesions were observed in six patients (including two with APSN). We observed intense expression of ANXA2 in glomerular and peritubular capillaries while expression of S100A10 was weaker. However, one of the patients with APSN showed strong S100A10 expression. Patients with LN and controls differed significantly in terms of S100A10 expression in peritubular capillaries. We also observed a statistical difference between patients who had LN with renal vascular lesions and those without renal vascular lesions in terms of ANXA2 expression in peritubular capillaries. CONCLUSIONS The presence of vascular lesions in LN appears to be associated with significant differences in the vascular expression of ANXA2. Vascular expression of ANXA2 was somewhat higher in LN. Vascular expression of S100A10 was somewhat lower in LN (except one of the two patients with APSN). Further studies of ANXA2's putative value as a biomarker of active LN or of vascular lesions in LN are required.
Collapse
Affiliation(s)
- V Salle
- Department of Internal Medicine, Amiens University Hospital, Amiens, France
| | - C Cordonnier
- Department of Pathology, Amiens University Hospital, Amiens, France
| | - J Schmidt
- Department of Internal Medicine, Amiens University Hospital, Amiens, France
| | - C Mazière
- INSERM U1088 Biochemistry Laboratory, Amiens University Hospital, Amiens, France
| | - A Smail
- Department of Internal Medicine, Amiens University Hospital, Amiens, France
| | - C Attencourt
- Department of Pathology, Amiens University Hospital, Amiens, France
| | - M P Mabille
- Department of Pathology, Amiens University Hospital, Amiens, France
| | - J C Mazière
- INSERM U1088 Biochemistry Laboratory, Amiens University Hospital, Amiens, France
| | - R Makdassi
- Department of Nephrology, Amiens University Hospital, Amiens, France
| | - G Choukroun
- Department of Nephrology, Amiens University Hospital, Amiens, France
| | - M Diouf
- Division of Clinical Research and Innovation, Amiens University Hospital, Amiens, France
| | - P Duhaut
- Department of Internal Medicine, Amiens University Hospital, Amiens, France
| | - J P Ducroix
- Department of Internal Medicine, Amiens University Hospital, Amiens, France
| |
Collapse
|
46
|
Abstract
Eukaryotic cells have been confronted throughout their evolution with potentially lethal plasma membrane injuries, including those caused by osmotic stress, by infection from bacterial toxins and parasites, and by mechanical and ischemic stress. The wounded cell can survive if a rapid repair response is mounted that restores boundary integrity. Calcium has been identified as the key trigger to activate an effective membrane repair response that utilizes exocytosis and endocytosis to repair a membrane tear, or remove a membrane pore. We here review what is known about the cellular and molecular mechanisms of membrane repair, with particular emphasis on the relevance of repair as it relates to disease pathologies. Collective evidence reveals membrane repair employs primitive yet robust molecular machinery, such as vesicle fusion and contractile rings, processes evolutionarily honed for simplicity and success. Yet to be fully understood is whether core membrane repair machinery exists in all cells, or whether evolutionary adaptation has resulted in multiple compensatory repair pathways that specialize in different tissues and cells within our body.
Collapse
Affiliation(s)
- Sandra T Cooper
- Institute for Neuroscience and Muscle Research, Kids Research Institute, The Children's Hospital at Westmead, Sydney, New South Wales, Australia; Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia; and Department of Cellular Biology and Anatomy, Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, Georgia
| | - Paul L McNeil
- Institute for Neuroscience and Muscle Research, Kids Research Institute, The Children's Hospital at Westmead, Sydney, New South Wales, Australia; Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, New South Wales, Australia; and Department of Cellular Biology and Anatomy, Institute of Molecular Medicine and Genetics, Georgia Regents University, Augusta, Georgia
| |
Collapse
|
47
|
Jiang Y, Fan X, Yu Z, Liao Z, Wang XS, van Leyen K, Sun X, Lo EH, Wang X. Combination Low-Dose Tissue-Type Plasminogen Activator Plus Annexin A2 for Improving Thrombolytic Stroke Therapy. Front Cell Neurosci 2015; 9:397. [PMID: 26528130 PMCID: PMC4604305 DOI: 10.3389/fncel.2015.00397] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/22/2015] [Indexed: 01/14/2023] Open
Abstract
Risk of hemorrhagic transformation, incomplete reperfusion, neurotoxicity, and a short treatment time window comprises major challenges for tissue plasminogen activator (tPA) thrombolytic stroke therapy. Improving tPA therapy has become one of the highest priorities in the stroke field. This mini review article focuses on our recent efforts aimed at evaluating a novel combination approach of low-dose tPA plus recombinant annexin A2 (rA2, a tPA, and plasminogen co-receptor), which might enhance tPA thrombolytic efficacy, while reducing its associated complications related to intracerebral hemorrhagic transformation. Results of our experimental studies using a focal embolic stroke model in rats support the feasibility of the combination approach and suggest the potential for successful clinical translation.
Collapse
Affiliation(s)
- Yinghua Jiang
- Department of Neurosurgery, The First Affiliated Hospital, Chongqing Medical University , Chongqing , China ; Neuroscience Program, Neuroprotection Research Laboratory, Department of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School , Boston, MA , USA
| | - Xiang Fan
- Neuroscience Program, Neuroprotection Research Laboratory, Department of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School , Boston, MA , USA ; Research Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine , Tianjin , China
| | - Zhanyang Yu
- Neuroscience Program, Neuroprotection Research Laboratory, Department of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School , Boston, MA , USA
| | - Zhengbu Liao
- Department of Neurosurgery, The First Affiliated Hospital, Chongqing Medical University , Chongqing , China ; Neuroscience Program, Neuroprotection Research Laboratory, Department of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School , Boston, MA , USA
| | - Xiao-Shu Wang
- Department of Neurosurgery, The First Affiliated Hospital, Chongqing Medical University , Chongqing , China ; Neuroscience Program, Neuroprotection Research Laboratory, Department of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School , Boston, MA , USA
| | - Klaus van Leyen
- Neuroscience Program, Neuroprotection Research Laboratory, Department of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School , Boston, MA , USA
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital, Chongqing Medical University , Chongqing , China
| | - Eng H Lo
- Neuroscience Program, Neuroprotection Research Laboratory, Department of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School , Boston, MA , USA
| | - Xiaoying Wang
- Neuroscience Program, Neuroprotection Research Laboratory, Department of Neurology and Radiology, Massachusetts General Hospital, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
48
|
Shetty P, Bargale A, Patil BR, Mohan R, Dinesh US, Vishwanatha JK, Gai PB, Patil VS, Amsavardani TS. Cell surface interaction of annexin A2 and galectin-3 modulates epidermal growth factor receptor signaling in Her-2 negative breast cancer cells. Mol Cell Biochem 2015; 411:221-33. [PMID: 26438086 DOI: 10.1007/s11010-015-2584-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/26/2015] [Indexed: 10/23/2022]
Abstract
Overexpression and activation of tyrosine kinase receptors like EGFR and Src regulate the progression and metastasis of Her-2 negative breast cancer. Recently we have reported the role of cell membrane interaction of phospholipid-binding protein annexin A2 (AnxA2) and EGFR in regulating cellular signaling in the activation of angiogenesis, matrix degradation, invasion, and cancer metastasis. Beta-galactoside-specific animal lectin galectin-3 is an apoptosis inhibitor, and cell surface-associated extracellular galectin-3 also has a role in cell migration, cancer progression, and metastasis. Similar expression pattern and membrane co-localization of these two proteins made us to hypothesize in the current study that galectin-3 and AnxA2 interaction is critical for Her-2 negative breast cancer progression. By various experimental analyses, we confirm that glycosylated AnxA2 at the membrane surface interacts with galectin-3. N-linked glycosylation inhibitor tunicamycin treatment convincingly blocked AnxA2 membrane translocation and its association with galectin-3. To analyze whether this interaction has any functional relevance, we tried to dissociate this interaction with purified plant lectin from chickpea (Cicer arietinum agglutinin). This highly specific 30 kDa plant lectin could dissociate AnxA2 from endogenous lectin galectin-3 interaction at the cell surface. This dissociation could down-regulate Bcl-2 family proteins, cell proliferation, and migration simultaneously triggering cell apoptosis. Targeting this interaction of membrane surface glycoprotein and its animal lectin in Her-2 negative breast cancer may be of therapeutic value.
Collapse
Affiliation(s)
- Praveenkumar Shetty
- Central Research Laboratory, SDM College of Medical Sciences & Hospital, Manjushree Nagar, Sattur, Dharwad, 580 009, India. .,Department of Biochemistry, SDM College of Medical Sciences & Hospital, Manjushree Nagar, Sattur, Dharwad, 580 009, India.
| | - Anil Bargale
- Central Research Laboratory, SDM College of Medical Sciences & Hospital, Manjushree Nagar, Sattur, Dharwad, 580 009, India.,Department of Biochemistry, SDM College of Medical Sciences & Hospital, Manjushree Nagar, Sattur, Dharwad, 580 009, India
| | | | - Rajashekar Mohan
- Department of Surgery, SDM College of Medical Sciences & Hospital, Dharwad, India
| | - U S Dinesh
- Department of Pathology, SDM College of Medical Sciences & Hospital, Dharwad, India
| | - Jamboor K Vishwanatha
- Department of Molecular Medicine, University of North Texas Health Science Center at Fort Worth, Fort Worth, TX, USA
| | - Pramod B Gai
- Karnataka Institute of DNA Research, Dharwad, India
| | - Vidya S Patil
- Central Research Laboratory, SDM College of Medical Sciences & Hospital, Manjushree Nagar, Sattur, Dharwad, 580 009, India.,Department of Biochemistry, SDM College of Medical Sciences & Hospital, Manjushree Nagar, Sattur, Dharwad, 580 009, India
| | - T S Amsavardani
- Department of Oral Pathology, Indira Gandhi Institute of Dental Sciences, Pondicherry, India
| |
Collapse
|
49
|
Xu XH, Pan W, Kang LH, Feng H, Song YQ. Association of annexin A2 with cancer development (Review). Oncol Rep 2015; 33:2121-8. [PMID: 25760910 DOI: 10.3892/or.2015.3837] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 02/20/2015] [Indexed: 01/11/2023] Open
Abstract
Annexin A2 (ANXA2) is a well-known calcium-dependent phospholipid binding protein widely distributed in the nucleus, cytoplasm and extracellular surface of various eukaryotic cells. It has been recognized as a pleiotropic protein affecting a wide range of molecular and cellular processes. Dysregulation and abnormal expression of ANXA2 are linked to a large number of prevalent diseases, including autoimmune and neurodegenerative disease, antiphospholipid syndrome, inflammation, diabetes mellitus and a series of cancers. Accumulating data suggest that ANXA2 is aberrantly expressed in a wide spectrum of cancers, and exerts profound effects on tumor cell adhesion, proliferation, apoptosis, invasion and metastasis as well as tumor neovascularization via different modes of action. However, despite significant research, our knowledge of the mechanism by which ANXA2 participates in cancer development remains fragmented. The present review systematically summarizes the effects of ANXA2 on tumor progression, in an attempt to gain an improved understanding of the underlying mechanisms and to provide a potential effective target for cancer therapy.
Collapse
Affiliation(s)
- Xiao-Heng Xu
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Wei Pan
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Li-Hua Kang
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Hui Feng
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, P.R. China
| | - Yan-Qiu Song
- Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, P.R. China
| |
Collapse
|
50
|
Jung H, Kim JS, Kim WK, Oh KJ, Kim JM, Lee HJ, Han BS, Kim DS, Seo YS, Lee SC, Park SG, Bae KH. Intracellular annexin A2 regulates NF-κB signaling by binding to the p50 subunit: implications for gemcitabine resistance in pancreatic cancer. Cell Death Dis 2015; 6:e1606. [PMID: 25611381 PMCID: PMC4669756 DOI: 10.1038/cddis.2014.558] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 11/26/2014] [Accepted: 11/26/2014] [Indexed: 12/18/2022]
Abstract
Annexin A2 (ANXA2) expression is highly upregulated in many types of cancer. Although cell surface localization of ANXA2 has been reported to have a critical role in the progression and metastasis of a variety of tumors, including pancreatic cancer, the biological role of intracellular ANXA2 is not fully understood. Herein the role of intracellular ANXA2 was investigated in a pancreatic cancer cell line. We first determined whether ANXA2 is involved in NF-κB signaling pathways. ANXA2 bound to the p50 subunit of NF-κB in a calcium-independent manner, and the ANXA2-p50 complex translocated into the nucleus. Furthermore, ANXA2 increased the transcriptional activity of NF-κB in both the resting and activated states and upregulated the transcription of several target genes downstream of NF-κB, including that encoding interleukin (IL)-6, which contributes to anti-apoptotic signaling. In Mia-Paca2 cells, we determined the effects of wild-type ANXA2 and an ANXA2 mutant, Y23A, which suppresses the cell surface localization, on upregulation of NF-κB transcriptional activity and secretion of IL-6. Both wild-type and Y23A ANXA2 induced anti-apoptotic effects in response to treatment with tumor necrosis factor-α or gemcitabine. Based on these results, we suggest that ANXA2 mediates resistance to gemcitabine by directly increasing the activity of NF-κB. Collectively, these data may provide additional information about the biological role of ANXA2 in pancreatic cancer and suggest that ANXA2 is a potential biomarker for the drug resistance phenotype and a candidate therapeutic target for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- H Jung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 305-701, Republic of Korea
- Functional Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 305-806, Republic of Korea
| | - J S Kim
- Functional Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 305-806, Republic of Korea
| | - W K Kim
- Functional Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 305-806, Republic of Korea
| | - K-J Oh
- Functional Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 305-806, Republic of Korea
| | - J-M Kim
- Department of Pathology, College of Medicine, Chungnam National University, Daejeon, 301-721, Republic of Korea
| | - H J Lee
- Department of Internal Medicine, Chungnam National University, Daejeon, 301-721, Republic of Korea
| | - B S Han
- Functional Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 305-806, Republic of Korea
| | - D S Kim
- Department of Pathology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 135-710, Republic of Korea
| | - Y S Seo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 305-701, Republic of Korea
| | - S C Lee
- Functional Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 305-806, Republic of Korea
| | - S G Park
- Functional Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 305-806, Republic of Korea
| | - K-H Bae
- Functional Genomics Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 305-806, Republic of Korea
| |
Collapse
|