1
|
Tao J, Yin L, Wu A, Zhang J, Zhang J, Shi H, Liu S, Niu L, Xu L, Feng Y, Lian S, Li L, Zeng L, Meng X, Zhou X, Liu T, Zhang L. PDIA2 Bridges Endoplasmic Reticulum Stress and Metabolic Reprogramming During Malignant Transformation of Chronic Colitis. Front Oncol 2022; 12:836087. [PMID: 35860571 PMCID: PMC9289542 DOI: 10.3389/fonc.2022.836087] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 06/02/2022] [Indexed: 12/03/2022] Open
Abstract
Background Chronic inflammation contributes to approximately 20% of cancers; the underlying mechanisms are still elusive. Here, using an animal model of colitis to colon-cancerous transformation, we demonstrated that endoplasmic reticulum (ER) stress couples with metabolic reprogramming to promote a malignant transformation of chronic inflammation. Methods The animal model for chronic colitis to colon-cancerous transformation was established in C57BL/6N mice by azoxymethane (AOM) and dextran sodium sulfate (DSS) treatments. The differential proteins in control and AOM/DSS-treated colon mucosa were determined using proteomic analysis; the kinetics of metabolic modifications were monitored by mitochondrial oxygen flux, extracellular acidification, and targeted metabolomics; the molecule linker between ER stress and metabolic modifications were identified by coimmunoprecipitation, KEGG pathway analysis, and the subcutaneous tumor model using gene-specific knockdown colon cancer cells. Tissue array analysis were used to evaluate the differential protein in cancer and cancer-adjacent tissues. Results AOM/DSS treatment induced 38 tumors in 10 mice at the 14th week with the mean tumor size 9.35 ± 3.87 mm2, which was significantly decreased to 5.85 ± 0.95 mm2 by the ER stress inhibitor 4-phenylbutyric acid (4PBA). Seven differential proteins were determined from control (1,067 ± 48) and AOM/DSS-treated mucosa (1,077 ± 59); the level of ER protein PDIA2 (protein disulfide isomerase-associated 2) was increased over 7-fold in response to AOM/DSS treatment. PDIA2 interacted with 420 proteins that were involved in 8 signaling pathways, in particular with 53 proteins in metabolic pathways. PDIA2 translocated from ER to mitochondria and interacted with the components of complexes I and II to inhibit oxophosphorylation but increase glycolysis. Knockdown PDIA2 in colon cancer cells restored the metabolic imbalance and significantly repressed tumor growth in the xenograft animal model. 4PBA therapy inhibited the AOM/DSS-mediated overexpression of PDIA2 and metabolic modifications and suppressed colon cancer growth. In clinic, PDIA2 was overexpressed in colon cancer tissues rather than cancer-adjacent tissues and was related with the late stages and lymph node metastasis of colon cancer. Conclusions Persistent ER stress reprograms the metabolism to promote the malignant transformation of chronic colitis; PDIA2 serves as a molecule linker between ER stress and metabolic reprogramming. The inhibition of ER stress restores metabolic homeostasis and attenuates the cancerous transformation of chronic inflammation.
Collapse
Affiliation(s)
- Jie Tao
- Scientific Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Lin Yin
- Scientific Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Ao Wu
- Scientific Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jiaoli Zhang
- Scientific Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Jingpu Zhang
- Scientific Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Huichun Shi
- Scientific Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Siyuan Liu
- The College of Information, Mechanical and Electrical Engineering, Shanghai Normal University, Shanghai, China
| | - Liangfei Niu
- Scientific Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Li Xu
- Scientific Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Yanling Feng
- Clinical Pathology Laboratory, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Shixian Lian
- Department of Surgery, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Lei Li
- Department of Surgery, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Liyan Zeng
- Department of Clinical Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xianmin Meng
- Department of Clinical Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xiaohui Zhou
- Animal Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Tiefu Liu
- Scientific Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- *Correspondence: Lijun Zhang, ; Tiefu Liu,
| | - Lijun Zhang
- Scientific Research Institute, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- Department of Clinical Research Center, Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
- *Correspondence: Lijun Zhang, ; Tiefu Liu,
| |
Collapse
|
2
|
Cordido A, Vizoso-Gonzalez M, Nuñez-Gonzalez L, Molares-Vila A, Chantada-Vazquez MDP, Bravo SB, Garcia-Gonzalez MA. Quantitative Proteomic Study Unmasks Fibrinogen Pathway in Polycystic Liver Disease. Biomedicines 2022; 10:290. [PMID: 35203500 PMCID: PMC8869147 DOI: 10.3390/biomedicines10020290] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 02/05/2023] Open
Abstract
(1) Background: Polycystic liver disease (PLD) is a heterogeneous group of congenital disorders characterized by bile duct dilatation and cyst development derived from cholangiocytes. Nevertheless, the cystogenesis mechanism is currently unknown and the PLD treatment is limited to liver transplantation. Novel and efficient therapeutic approaches are th6us needed. In this context, the present work has a principal aim to find novel molecular pathways, as well as new therapeutic targets, involved in the hepatic cystogenesis process. (2) Methods: Quantitative proteomics based on SWATH-MS technology were performed comparing hepatic proteomes of Wild Type and mutant/polycystic livers in a polycystic kidney disease (PKD) murine model (Pkd1cond/cond;Tam-Cre-/+). (3) Results: We identified several proteins altered in abundance, with two-fold cut-off up-regulation or down-regulation and an adjusted p-value significantly related to hepatic cystogenesis. Then, we performed enrichment and a protein-protein analysis identifying a cluster focused on hepatic fibrinogens. Finally, we validated a selection of targets by RT-qPCR, Western blotting and immunohistochemistry, finding a high correlation with quantitative proteomics data and validating the fibrinogen complex. (4) Conclusions: This work identified a novel molecular pathway in cystic liver disease, highlighting the fibrinogen complex as a possible new therapeutic target for PLD.
Collapse
Affiliation(s)
- Adrian Cordido
- Group of Genetics and Developmental Biology of Renal Diseases, Nephrology Laboratory (N°11), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain; (A.C.); (M.V.-G.); (L.N.-G.)
- Genomic Medicine Group, Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain
| | - Marta Vizoso-Gonzalez
- Group of Genetics and Developmental Biology of Renal Diseases, Nephrology Laboratory (N°11), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain; (A.C.); (M.V.-G.); (L.N.-G.)
- Genomic Medicine Group, Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain
| | - Laura Nuñez-Gonzalez
- Group of Genetics and Developmental Biology of Renal Diseases, Nephrology Laboratory (N°11), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain; (A.C.); (M.V.-G.); (L.N.-G.)
- Genomic Medicine Group, Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain
| | - Alberto Molares-Vila
- Biostatistics Platform, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain;
| | - Maria del Pilar Chantada-Vazquez
- Proteomic Platform, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain;
| | - Susana B. Bravo
- Proteomic Platform, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain;
| | - Miguel A. Garcia-Gonzalez
- Group of Genetics and Developmental Biology of Renal Diseases, Nephrology Laboratory (N°11), Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain; (A.C.); (M.V.-G.); (L.N.-G.)
- Genomic Medicine Group, Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain
- Galician Public Foundation of Genomic Medicine, Santiago de Compostela Clinical Hospital Complex (CHUS), 15706 Santiago de Compostela, Spain
| |
Collapse
|
3
|
Lim HI, Hajjar KA. Annexin A2 in Fibrinolysis, Inflammation and Fibrosis. Int J Mol Sci 2021; 22:6836. [PMID: 34202091 PMCID: PMC8268605 DOI: 10.3390/ijms22136836] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/12/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
As a cell surface tissue plasminogen activator (tPA)-plasminogen receptor, the annexin A2 (A2) complex facilitates plasmin generation on the endothelial cell surface, and is an established regulator of hemostasis. Whereas A2 is overexpressed in hemorrhagic disease such as acute promyelocytic leukemia, its underexpression or impairment may result in thrombosis, as in antiphospholipid syndrome, venous thromboembolism, or atherosclerosis. Within immune response cells, A2 orchestrates membrane repair, vesicle fusion, and cytoskeletal organization, thus playing a critical role in inflammatory response and tissue injury. Dysregulation of A2 is evident in multiple human disorders, and may contribute to the pathogenesis of various inflammatory disorders. The fibrinolytic system, moreover, is central to wound healing through its ability to remodel the provisional matrix and promote angiogenesis. A2 dysfunction may also promote tissue fibrogenesis and end-organ fibrosis.
Collapse
Affiliation(s)
- Hana I. Lim
- Division of Hematology and Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA;
| | - Katherine A. Hajjar
- Division of Hematology and Oncology, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
4
|
Qiu LW, Liu YF, Cao XQ, Wang Y, Cui XH, Ye X, Huang SW, Xie HJ, Zhang HJ. Annexin A2 promotion of hepatocellular carcinoma tumorigenesis via the immune microenvironment. World J Gastroenterol 2020; 26:2126-2137. [PMID: 32476780 PMCID: PMC7235202 DOI: 10.3748/wjg.v26.i18.2126] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/08/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer with a dismal prognosis, especially when diagnosed at advanced stages. Annexin A2 (ANXA2), is found to promote cancer progression and therapeutic resistance. However, the underlining mechanisms of ANXA2 in immune escape of HCC remain poorly understood up to now. Herein, we summarized the molecular function of ANXA2 in HCC and its relationship with prognosis. Furthermore, we tentatively elucidated the underlying mechanism of ANXA2 immune escape of HCC by upregulating the proportion of regulatory T cells and the expression of several inhibitory molecules, and by downregulating the proportion of natural killer cells and dendritic cells and the expression of several inhibitory molecules or effector molecules. We expect a lot of in-depth studies to further reveal the underlying mechanism of ANXA2 in immune escape of HCC in the future.
Collapse
Affiliation(s)
- Li-Wei Qiu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Yi-Fei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Xiao-Qing Cao
- Department of Thoracic Surgery, Beijing Chest Hospital, Capital Medical University (Beijing Tuberculosis and Thoracic Tumor Research Institute), Beijing 101149, China
| | - Yan Wang
- Emergency Department, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Xiao-Hong Cui
- Department of General Surgery, Shanghai Electric Power Hospital, Shanghai 200050, China
| | - Xian Ye
- Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Shuo-Wen Huang
- Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Hong-Jun Xie
- Medical School of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Hai-Jian Zhang
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| |
Collapse
|
5
|
Zhang Y, Shen Y, Yin L, Qi T, Jia X, Lu H, Zhang L. Plasma Membrane Proteomic Profile Discovers Macrophage-capping Protein Related to Latent HIV-1. Curr HIV Res 2020; 17:42-52. [PMID: 31057110 DOI: 10.2174/1570162x17666190506155222] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/24/2019] [Accepted: 04/25/2019] [Indexed: 02/03/2023]
Abstract
BACKGROUND Due to the persistence of latent HIV-infected cellular reservoirs, HIV virus can not be eradicated completely. OBJECTIVE To identify proteins related to HIV latency, we performed a subcellular proteomic study in HIV latent cell lines. METHODS An established HIV-1 latent cell model (J-Lat Tat-GFP Clone A7 cells, A7 cells) and its parental cell line (Jurkat cells) were used. The plasma membrane (PM) fraction from cultured cells was enriched through aqueous two-phase partition. PM proteins were extracted and then separated using two-dimensional electrophoresis (2DE). Differentially expressed proteins were identified by mass spectrometry, and verified by western blotting. RESULTS Thirteen non-redundant proteins were identified to be differentially expressed in the A7 PM fraction compared to those in the Jurkat PM. Eight had a PM location through Gene Ontology (GO) analysis. A differential protein network of CAPG-ACTR3-CD3D was detected to have interactions with HIV Vpr, Tat, gp160, etc. through STRING software analysis. One of the differential proteins (Macrophage-capping protein (CAPG)) was verified by western blotting to be down- regulated in two cell lines and HIV resting CD4+ T cells negatively selected from patients. CONCLUSION We identified 13 proteins in A7 compared to Jurkat cells. CAPG may be a potential biomarker related to HIV latency.
Collapse
Affiliation(s)
- Yujiao Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Yinzhong Shen
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Lin Yin
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Tangkai Qi
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Xiaofang Jia
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Hongzhou Lu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Lijun Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| |
Collapse
|
6
|
Nuerzhati Y, Dong R, Song Z, Zheng S. Role of the long non‑coding RNA‑Annexin A2 pseudogene 3/Annexin A2 signaling pathway in biliary atresia‑associated hepatic injury. Int J Mol Med 2018; 43:739-748. [PMID: 30569159 PMCID: PMC6317672 DOI: 10.3892/ijmm.2018.4023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Accepted: 11/21/2018] [Indexed: 12/29/2022] Open
Abstract
Biliary atresia (BA) is the most common cause of chronic cholestasis in children. The long non‑coding RNA (lncRNA) Annexin A2 pseudogene 3 (ANXA2P3) and Annexin A2 (ANXA2) have been suggested to serve pivotal roles in BA; however, the clinical significance and biological roles of ANXA2P3 and ANXA2 in BA remain to be elucidated. The present study aimed to elucidate the function of ANAX2P3 and ANXA2 in BA‑induced liver injury using a human liver cell line and liver tissues from patients with BA. Reverse transcription‑quantitative polymerase chain reaction, western blotting and immunohistochemistry were conducted to determine the expression levels of ANXA2 and ANXA2P3 in liver tissues from patients with BA. Classification of fibrosis was analyzed by Masson staining. The functional roles of ANXA2 and ANXA2P3 in liver cells were determined by Cell Counting kit‑8 assay, and flow cytometric and cell cycle analyses. Activation of the ANXA2/ANXA2P3 signaling pathway in liver cells was evaluated by western blot analysis. According to the present results, the expression levels of ANXA2 and ANXA2P3 were significantly increased in liver tissues from patients with BA. In addition, knocking down the expression of ANXA2P3 and ANXA2 may result in reduced liver cell proliferation, cell cycle arrest in G1 phase and increased apoptosis of liver cells in vitro. Furthermore, in cells in which ANXA2 and ANXA2P3 were overexpressed, cell apoptosis was reduced and cell cycle arrest in G2 phase. Taken together, these results indicated that ANXA2P3 and ANXA2 may have protective effects against liver injury progression and may be considered biomarkers in patients with BA.
Collapse
Affiliation(s)
- Yeletai Nuerzhati
- Department of Pediatric Hepatobiliary Surgery, Children's Hospital of Fudan University, Shanghai 201102, P.R. China
| | - Rui Dong
- Department of Pediatric Hepatobiliary Surgery, Children's Hospital of Fudan University, Shanghai 201102, P.R. China
| | - Zai Song
- Department of Pediatric Hepatobiliary Surgery, Children's Hospital of Fudan University, Shanghai 201102, P.R. China
| | - Shan Zheng
- Department of Pediatric Hepatobiliary Surgery, Children's Hospital of Fudan University, Shanghai 201102, P.R. China
| |
Collapse
|
7
|
Reciprocal regulation of pro-inflammatory Annexin A2 and anti-inflammatory Annexin A1 in the pathogenesis of rheumatoid arthritis. Mol Biol Rep 2018; 46:83-95. [PMID: 30426384 DOI: 10.1007/s11033-018-4448-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 10/16/2018] [Indexed: 02/06/2023]
Abstract
Annexin A2 has been implicated in several immune modulated diseases including Rheumatoid arthritis (RA) pannus formation. The most relied treatment option for RA pathogenesis is glucocorticoids. Glucocorticoids regulate the synthesis, phosphorylation and cellular deposition of Annexin A1. This annexin mediates the anti-inflammatory actions of glucocorticoids. These two first characterized members of annexin superfamily proteins acts reciprocally, one as an anti-inflammatory and the other proinflammatory in nature. The possibility of these molecules as soluble biomarkers and as an upstream regulator of major cytokine devastation at RA microenvironment has not been previously explored. Current study elucidates the reciprocal regulation of these two annexins in RA pathogenesis. These Annexin A2/A1 and downstream cytokines in RA serum were analysed by ELISA. Western blot, Immunocytochemistry, immunoprecipitation and Immunohistochemistry were adapted to analyse these molecules in tissue and synovial fibroblasts and also in different experimental conditions. Significant increase in the level of Annexin A2 was noticed in naïve RA patients compared to controls (14.582 ± 1.766 ng/ml vs. 7.37 ± 1.450 ng/ml; p ≤ 0.001). In remission cases significant low levels was detected. On the contrary, significant decrease in the level of Annexin A1 was noticed in naïve RA patients compared to healthy controls (12.322 ± 2.91 vs. 16.998 ± 4.298 ng/ml; p ≤ 0.001), wherein remission cases serum Annexin A1 was significantly high. The knockdown of proinflammatory Annexin A2 by siRNA/antibody treatment could mimic the glucocorticoid treatment as which induced cellular Annexin A1 and membrane translocation resulting in the terminal action. Current data elucidating the regulatory interplay between Annexin A2 and Annexin A1 in RA pathogenesis.
Collapse
|
8
|
Amhimmid Badr S, Waheeb Fahmi M, Mahmoud Nomir M, Mohammad El-Shishtawy M. Calcium channel α2δ1 subunit as a novel biomarker for diagnosis of hepatocellular carcinoma. Cancer Biol Med 2018; 15:52-60. [PMID: 29545968 PMCID: PMC5842334 DOI: 10.20892/j.issn.2095-3941.2017.0167] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Objective: Hepatocellular carcinoma (HCC) is the fifth most common malignancy worldwide. The identification of new simple, inexpensive and highly accurate markers for HCC diagnosis and screening is needed. This case-control study evaluates the role of annexin A2 and voltage-gated calcium channels α2δ1 subunit as serum biomarkers for HCC diagnosis. Methods: The study comprised three groups: group 1, 50 patients with an initial diagnosis of HCC associated with chronic hepatitis C virus infection; group 2, 25 patients diagnosed with chronic hepatitis C virus infection and cirrhosis without any evidence of HCC; and group 3, 15 healthy controls. All participants were subjected to clinical and laboratory investigations, and radiological scanning. The serum levels of alpha-fetoprotein (AFP), annexin A2, and the α2δ1 subunit were evaluated by using ELISA technique. Results: The serum levels of annexin A2 significantly increased in patients with HCC (10.4±2.5 ng/mL; P<0.001) or with cirrhosis (9.31±1.8 ng/mL;P<0.001) comparing to that of healthy controls (0.296±0.09 ng/mL). However, there was no significant difference in serum annexin A2 levels in patients with HCC comparing to those with cirrhosis. Serum α2δ1 subunit significantly increased in patients with HCC (20.12±3.7 ng/mL) comparing to that in patients with cirrhosis (10.41±3.4 ng/mL,P<0.001) and healthy controls (10.2±2.9 ng/mL,P<0.001).
Conclusions: The serum α2δ1 subunit may function as a new biomarker for HCC diagnosis. Conversely, serum annexin A2 has low diagnostic value as an HCC marker, especially in patients with underlying cirrhosis.
Collapse
|
9
|
Kan F, Ye L, Yan T, Cao J, Zheng J, Li W. Proteomic and transcriptomic studies of HBV-associated liver fibrosis of an AAV-HBV-infected mouse model. BMC Genomics 2017; 18:641. [PMID: 28830339 PMCID: PMC5568174 DOI: 10.1186/s12864-017-3984-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 08/01/2017] [Indexed: 01/05/2023] Open
Abstract
Background Human hepatitis B virus (HBV) infection is an important public health issue in the Asia-Pacific region and is associated with chronic hepatitis, liver fibrosis, cirrhosis and even liver cancer. However, the underlying mechanisms of HBV-associated liver fibrosis remain incompletely understood. Results In the present study, proteomic and transcriptomic approaches as well as biological network analyses were performed to investigate the differentially expressed molecular signature and key regulatory networks that were associated with HBV-mediated liver fibrosis. RNA sequencing and 2DE-MALDI-TOF/TOF were performed on liver tissue samples obtained from HBV-infected C57BL/6 mouse generated via AAV8-HBV virus. The results showed that 322 genes and 173 proteins were differentially expressed, and 28 HBV-specific proteins were identified by comprehensive proteomic and transcriptomic analysis. GO analysis indicated that the differentially expressed proteins were predominantly involved in oxidative stress, which plays a key role in HBV-related liver fibrosis. Importantly, CAT, PRDX1, GSTP1, NXN and BLVRB were shown to be associated with oxidative stress among the differentially expressed proteins. The most striking results were validated by Western blot and RT-qPCR. The RIG-I like receptor signaling pathway was found to be the major signal pathway that changed during HBV-related fibrosis. Conclusions This study provides novel insights into HBV-associated liver fibrosis and reveals the significant role of oxidative stress in liver fibrosis. Furthermore, CAT, BLVRB, NXN, PRDX1, and IDH1 may be candidates for detection of liver fibrosis or therapeutic targets for the treatment of liver fibrosis. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3984-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fangming Kan
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lei Ye
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tao Yan
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jiaqi Cao
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianhua Zheng
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Wuping Li
- MOH Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
10
|
Wang Y, Terrell AM, Riggio BA, Anand D, Lachke SA, Duncan MK. β1-Integrin Deletion From the Lens Activates Cellular Stress Responses Leading to Apoptosis and Fibrosis. Invest Ophthalmol Vis Sci 2017; 58:3896-3922. [PMID: 28763805 PMCID: PMC5539801 DOI: 10.1167/iovs.17-21721] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 05/30/2017] [Indexed: 12/18/2022] Open
Abstract
Purpose Previous research showed that the absence of β1-integrin from the mouse lens after embryonic day (E) 13.5 (β1MLR10) leads to the perinatal apoptosis of lens epithelial cells (LECs) resulting in severe microphthalmia. This study focuses on elucidating the molecular connections between β1-integrin deletion and this phenotype. Methods RNA sequencing was performed to identify differentially regulated genes (DRGs) in β1MLR10 lenses at E15.5. By using bioinformatics analysis and literature searching, Egr1 (early growth response 1) was selected for further study. The activation status of certain signaling pathways (focal adhesion kinase [FAK]/Erk, TGF-β, and Akt signaling) was studied via Western blot and immunohistochemistry. Mice lacking both β1-integrin and Egr1 genes from the lenses were created (β1MLR10/Egr1-/-) to study their relationship. Results RNA sequencing identified 120 DRGs that include candidates involved in the cellular stress response, fibrosis, and/or apoptosis. Egr1 was investigated in detail, as it mediates cellular stress responses in various cell types, and is recognized as an upstream regulator of numerous other β1MLR10 lens DRGs. In β1MLR10 mice, Egr1 levels are elevated shortly after β1-integrin loss from the lens. Further, pErk1/2 and pAkt are elevated in β1MLR10 LECs, thus providing the potential signaling mechanism that causes Egr1 upregulation in the mutant. Indeed, deletion of Egr1 from β1MLR10 lenses partially rescues the microphthalmia phenotype. Conclusions β1-integrin regulates the appropriate levels of Erk1/2 and Akt phosphorylation in LECs, whereas its deficiency results in the overexpression of Egr1, culminating in reduced cell survival. These findings provide insight into the molecular mechanism underlying the microphthalmia observed in β1MLR10 mice.
Collapse
Affiliation(s)
- Yichen Wang
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
| | - Anne M. Terrell
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
| | - Brittany A. Riggio
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
| | - Deepti Anand
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
| | - Salil A. Lachke
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
| | - Melinda K. Duncan
- Department of Biological Sciences, University of Delaware, Newark, Delaware, United States
| |
Collapse
|
11
|
Schuliga M, Jaffar J, Berhan A, Langenbach S, Harris T, Waters D, Lee PVS, Grainge C, Westall G, Knight D, Stewart AG. Annexin A2 contributes to lung injury and fibrosis by augmenting factor Xa fibrogenic activity. Am J Physiol Lung Cell Mol Physiol 2017; 312:L772-L782. [DOI: 10.1152/ajplung.00553.2016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/06/2017] [Accepted: 03/06/2017] [Indexed: 12/11/2022] Open
Abstract
In lung injury and disease, including idiopathic pulmonary fibrosis (IPF), extravascular factor X is converted into factor Xa (FXa), a coagulant protease with fibrogenic actions. Extracellular annexin A2 binds to FXa, augmenting activation of the protease-activated receptor-1 (PAR-1). In this study, the contribution of annexin A2 in lung injury and fibrosis was investigated. Annexin A2 immunoreactivity was observed in regions of fibrosis, including those associated with fibroblasts in lung tissue of IPF patients. Furthermore, annexin A2 was detected in the conditioned media and an EGTA membrane wash of human lung fibroblast (LF) cultures. Incubation with human plasma (5% vol/vol) or purified FXa (15–50 nM) evoked fibrogenic responses in LF cultures, with FXa increasing interleukin-6 (IL-6) production and cell number by 270 and 46%, respectively ( P < 0.05, n = 5–8). The fibrogenic actions of plasma or FXa were attenuated by the selective FXa inhibitor apixaban (10 μM, or antibodies raised against annexin A2 or PAR-1 (2 μg/ml). FXa-stimulated LFs from IPF patients ( n = 6) produced twice as much IL-6 as controls ( n = 10) ( P < 0.05), corresponding with increased levels of extracellular annexin A2. Annexin A2 gene deletion in mice reduced bleomycin-induced increases in bronchoalveolar lavage fluid (BALF) IL-6 levels and cell number (* P < 0.05; n = 4–12). Lung fibrogenic gene expression and dry weight were reduced by annexin A2 gene deletion, but lung levels of collagen were not. Our data suggest that annexin A2 contributes to lung injury and fibrotic disease by mediating the fibrogenic actions of FXa. Extracellular annexin A2 is a potential target for the treatment of IPF.
Collapse
Affiliation(s)
- Michael Schuliga
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Jade Jaffar
- Department of Allergy, Immunology, and Respiratory Medicine, Alfred Hospital, Prahran, Victoria, Australia
| | - Asres Berhan
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Shenna Langenbach
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - Trudi Harris
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| | - David Waters
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Peter V. S. Lee
- Department of Mechanical Engineering, University of Melbourne, Parkville, Victoria, Australia
| | - Christopher Grainge
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, New South Wales, Australia; and
| | - Glen Westall
- Department of Allergy, Immunology, and Respiratory Medicine, Alfred Hospital, Prahran, Victoria, Australia
| | - Darryl Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alastair G. Stewart
- Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
12
|
Liu J, Li M, Gong J, Han P, Wang Y, Li D, Tian D, Liao J. Knockdown of histidine-rich calcium-binding protein (HRC) suppresses liver fibrosis by inhibiting the activation of hepatic stellate cells. Biol Open 2017; 6:29-34. [PMID: 27881436 PMCID: PMC5278420 DOI: 10.1242/bio.019828] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The histidine-rich calcium-binding protein (HRC) is a regulator of Ca2+ homeostasis and it plays a significant role in hepatocellular carcinoma (HCC) progression. However, the relationship between HRC and liver fibrogenesis is still unknown. Our data demonstrates that HRC was upregulated in fibrotic liver and activated hepatic stellate cells (HSCs). TGF-β treatment increased α-SMA and HRC expression dose-dependently in HSCs. Repression of HRC reduced α-SMA, CTGF and collagen expression, and inhibited HSC proliferation and migration. In addition, we found that the anti-fibrosis effect of HRC knockdown was associated with endoplasmic reticulum (ER) stress. Silencing of HRC decreased the expression of ER stress and autophagy markers. Moreover, ER stress agonist thapsigargin (TG) enhanced, whereas ER stress antagonist 4-phenylbutyric acid (4-PBA) alleviated HSCs activation and autophagy. In conclusion, these data indicate that depletion of HRC inhibited HSC activation through the ER stress pathway, and HRC may be a potential regulator of liver fibrosis.
Collapse
Affiliation(s)
- Jingmei Liu
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Mengke Li
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.,Department of Gastroenterology, Zhoushan Hospital, Zhoushan 316000, China
| | - Jin Gong
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ping Han
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yunwu Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dongxiao Li
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Dean Tian
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiazhi Liao
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
13
|
Yuan Y, Hou X, Feng H, Liu R, Xu H, Gong W, Deng J, Sun C, Gao Y, Peng J, Wu Y, Li J, Fang C, Chen Q. Proteomic identification of cyclophilin A as a potential biomarker and therapeutic target in oral submucous fibrosis. Oncotarget 2016; 7:60348-60365. [PMID: 27533088 PMCID: PMC5312388 DOI: 10.18632/oncotarget.11254] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 07/18/2016] [Indexed: 02/05/2023] Open
Abstract
Oral submucous fibrosis (OSF) is a pre-cancerous lesion, which is characterized by fibrosis of the oral submucosa. Despite large body of studies focusing on this disease, the molecular mechanisms underlying the progression of OSF remained unclear. In this study, 2-DE-based proteomic approaches were employed to identify the differently expressed proteins between OSF and normal tissues. In total, 88 proteins were identified with altered expression levels, including CypA. Upregulation of CypA was further validated through immunohistochemistry staining combined with Q-PCR and western blot by using clinical samples. Statistical analyses reveal that CypA expression level is correlated to the progression of OSF. Finally, functional study reveals a pro-proliferative property of CypA in fibroblast cells by using multiple in vitro models. The present data suggest that CypA might be a potential biomarker and therapeutic target for OSF, and will lead to a better understanding of OSF pathogenesis.
Collapse
Affiliation(s)
- Yao Yuan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu China, 610041
| | - Xiaohui Hou
- Department of Endodontics, School & Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, Shanghai China, 200072
| | - Hui Feng
- Xiangya Stomatological Hospital, Central South University, Changsha, China, 410008
| | - Rui Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu China, 610041
| | - Hao Xu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu China, 610041
| | - Wang Gong
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu China, 610041
| | - Jing Deng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu China, 610041
| | - Chongkui Sun
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu China, 610041
| | - Yijun Gao
- Department of Stomatology, Second Xiangya Hospital, Central South University, Changsha, China, 410008
| | - Jieying Peng
- Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China, 410008
| | - Yingfang Wu
- Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China, 410008
| | - Jiang Li
- Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China, 410008
| | - Changyun Fang
- Center of Stomatology, Xiangya Hospital, Central South University, Changsha, China, 410008
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu China, 610041
| |
Collapse
|
14
|
Kolgelier S, Demir NA, Inkaya AC, Sumer S, Ozcimen S, Demir LS, Pehlivan FS, Arslan M, Arpaci A. Serum Levels of Annexin A2 as a Candidate Biomarker for Hepatic Fibrosis in Patients With Chronic Hepatitis B. HEPATITIS MONTHLY 2015; 15:e30655. [PMID: 26587036 PMCID: PMC4644632 DOI: 10.5812/hepatmon.30655] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 08/31/2015] [Accepted: 08/31/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND Hepatologists have studied serologic markers of liver injury for decades. Annexins are a prominent group of such markers and annexin A2 (AnxA2) is one of the best characterized annexins. AnxA2 inhibits HBV polymerase among other functions. Its expression is up-regulated in regenerative hepatocytes. OBJECTIVES To determine if serum AnxA2 level has a role in estimating liver damage in chronic HBV infection and investigate whether AnxA2 levels correlate with hepatic fibrosis. PATIENTS AND METHODS This study included 173 patients with chronic hepatitis B (CHB) and 51 healthy controls. Liver fibrosis was graded histologically on liver biopsy samples. Blood samples were taken from patients during biopsy and serum AnxA2 levels were measured with ELISA. RESULTS In a group of adult patients with CHB, AnxA2 values were far higher than those of the control group (P = 0.001). When we assessed AnxA2 levels based on fibrosis stages, serum AnxA2 levels of patients with early stage fibrosis (stages 1 - 3) were significantly higher than those of patients with advanced stage fibrosis (stages 4 - 5; P = 0.001). CONCLUSIONS AnxA2 is a useful biomarker for early stage fibrosis in patients with CHB.
Collapse
Affiliation(s)
- Servet Kolgelier
- Department of Infectious Diseases and Clinical Microbiology, Adiyaman University Faculty of Medicine, Adiyaman, Turkey
- Corresponding Author: Servet Kolgelier, Department of Infectious Diseases and Clinical Microbiology, Adiyaman University Faculty of Medicine, Adiyaman, Turkey. Tel: +90-5052671529, Fax: +90-3322412184, E-mail:
| | - Nazlim Aktug Demir
- Department of Infectious Diseases and Clinical Microbiology, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Ahmet Cagkan Inkaya
- Department of Infectious Diseases and Clinical Microbiology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Sua Sumer
- Department of Infectious Diseases and Clinical Microbiology, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Serap Ozcimen
- Department of Infectious Diseases and Clinical Microbiology, Konya State Hospital, Konya, Turkey
| | - Lutfi Saltuk Demir
- Department of Public Health, Faculty of Medicine, Necmettin Erbakan University, Konya, Turkey
| | | | - Mahmure Arslan
- Department of Biochemistry, Adiyaman State Hospital, Adiyaman, Turkey
| | - Abdullah Arpaci
- Department of Biochemistry, Faculty of Medicine, Adiyaman University, Adiyaman, Turkey
| |
Collapse
|
15
|
Pianta A, Drouin EE, Crowley JT, Arvikar S, Strle K, Costello CE, Steere AC. Annexin A2 is a target of autoimmune T and B cell responses associated with synovial fibroblast proliferation in patients with antibiotic-refractory Lyme arthritis. Clin Immunol 2015; 160:336-41. [PMID: 26187145 DOI: 10.1016/j.clim.2015.07.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 07/06/2015] [Indexed: 01/12/2023]
Abstract
In this study, autoantibody responses to annexin A2 were found in 11-15% of 278 patients with Lyme disease, including in those with erythema migrans (EM), an early sign of the illness, and in those with antibiotic-responsive or antibiotic-refractory Lyme arthritis (LA), a late disease manifestation. In contrast, robust T cell reactivity to annexin A2 peptides was found only in patients with responsive or refractory LA. In LA patients, annexin A2 protein levels, which were higher in the refractory group, correlated with annexin A2 antibody levels in sera and synovial fluid. In addition, in patients with antibiotic-refractory LA who had anti-annexin A2 antibodies, synovial tissue had intense staining for annexin A2 protein, greater synovial fibroblast proliferation and more tissue fibrosis. Thus, a subset of LA patients had T and B cell responses to annexin A2, and in the refractory group, annexin A2 autoantibodies were associated with specific pathologic findings.
Collapse
Affiliation(s)
- Annalisa Pianta
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, United States.
| | - Elise E Drouin
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - Jameson T Crowley
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - Sheila Arvikar
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - Klemen Strle
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| | - Catherine E Costello
- Center for Biomedical Mass Spectrometry, Boston University School of Medicine, Boston, United States
| | - Allen C Steere
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, United States
| |
Collapse
|
16
|
Lancha A, Rodríguez A, Catalán V, Becerril S, Sáinz N, Ramírez B, Burrell MA, Salvador J, Frühbeck G, Gómez-Ambrosi J. Osteopontin deletion prevents the development of obesity and hepatic steatosis via impaired adipose tissue matrix remodeling and reduced inflammation and fibrosis in adipose tissue and liver in mice. PLoS One 2014; 9:e98398. [PMID: 24871103 PMCID: PMC4037189 DOI: 10.1371/journal.pone.0098398] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 05/02/2014] [Indexed: 02/06/2023] Open
Abstract
Osteopontin (OPN) is a multifunctional extracellular matrix (ECM) protein involved in multiple physiological processes. OPN expression is dramatically increased in visceral adipose tissue in obesity and the lack of OPN protects against the development of insulin resistance and inflammation in mice. We sought to unravel the potential mechanisms involved in the beneficial effects of the absence of OPN. We analyzed the effect of the lack of OPN in the development of obesity and hepatic steatosis induced by a high-fat diet (HFD) using OPN-KO mice. OPN expression was upregulated in epididymal white adipose tissue (EWAT) and liver in wild type (WT) mice with HFD. OPN-KO mice had higher insulin sensitivity, lower body weight and fat mass with reduced adipose tissue ECM remodeling and reduced adipocyte size than WT mice under a HFD. Reduced MMP2 and MMP9 activity was involved in the decreased ECM remodeling. Crown-like structure number in EWAT as well as F4/80-positive cells and Emr1 expression in EWAT and liver increased with HFD, while OPN-deficiency blunted the increase. Moreover, our data show for the first time that OPN-KO under a HFD mice display reduced fibrosis in adipose tissue and liver, as well as reduced oxidative stress in adipose tissue. Gene expression of collagens Col1a1, Col6a1 and Col6a3 in EWAT and liver, as well as the profibrotic cytokine Tgfb1 in EWAT were increased with HFD, while OPN-deficiency prevented this increase. OPN deficiency prevented hepatic steatosis via reduction in the expression of molecules involved in the onset of fat accumulation such as Pparg, Srebf1, Fasn, Mogat1, Dgat2 and Cidec. Furthermore, OPN-KO mice exhibited higher body temperature and improved BAT function. The present data reveal novel mechanisms of OPN in the development of obesity, pointing out the inhibition of OPN as a promising target for the treatment of obesity and fatty liver.
Collapse
Affiliation(s)
- Andoni Lancha
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - Neira Sáinz
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
| | - María A. Burrell
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Histology and Pathology, University of Navarra, Pamplona, Spain
| | - Javier Salvador
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, Madrid, Spain
- * E-mail:
| |
Collapse
|
17
|
Liu Z, Ling Q, Wang J, Xie H, Xu X, Zheng S. Annexin A2 is not a good biomarker for hepatocellular carcinoma in cirrhosis. Oncol Lett 2013; 6:125-129. [PMID: 23946789 PMCID: PMC3742823 DOI: 10.3892/ol.2013.1337] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Accepted: 04/11/2013] [Indexed: 12/11/2022] Open
Abstract
In China, hepatocellular carcinoma (HCC) usually develops following a long history of chronic hepatitis B infection or cirrhosis. To evaluate the diagnostic role of annexin A2 (ANXA2), a possible tumor marker, in patients with hepatitis B virus (HBV)-related HCC, particularly those with a history of cirrhosis, the present study prospectively enrolled 87 patients with HBV-related HCC (with cirrhosis), 39 patients with HBV-related cirrhosis and 27 healthy controls. The expression levels of serum and tissue ANXA2 were determined using an enzyme-linked immunosorbent assay (ELISA) and immunohistochemical staining, respectively. The serum levels of ANXA2 were significantly elevated in the patients with HCC (median, 567.2 μg/ml; P=0.003) and cirrhosis (median, 414.8 μg/ml; P=0.011) compared with the healthy controls (median, 241.9 μg/ml). However, no significant differences were observed in the serum ANXA2 levels between the patients with HCC and those with cirrhosis. The immunohistochemical staining analysis showed that the healthy controls did not show positive staining, while the number of cases immunoreactive for ANXA2 steadily increased from the liver cirrhosis tissues (20/39, 51.3%) to the non-cancer (53/87, 60.9%) and cancer tissues (68/87, 78.2%). The cancer tissues exhibited a significantly higher ANXA2-positive rate compared with the non-cancer (P=0.013) and liver cirrhosis tissues (P=0.002). Furthermore, marked ANXA2 staining was more prevalent in the cancer tissues (16/87, 18.4%) than the non-cancer (4/87, 4.6%; P=0.004) and liver cirrhosis (1/39, 2.6%; P=0.034) tissues. The sensitivity, specificity and diagnostic accuracy of tissue ANXA2 for HCC in cirrhosis were 78.2, 42.1 and 56.8%, respectively. The ANXA2 expression levels in the serum and cancer tissues were not associated with tumor-free survival or patient survival following liver transplantation. Serum or tissue ANXA2 is not a good diagnostic marker for HCC in HBV-related cirrhosis and is not associated with prognosis.
Collapse
Affiliation(s)
- Zhikun Liu
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China ; Department of Surgery, Division of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | | | | | | | | | | |
Collapse
|
18
|
Jia X, Yin L, Feng Y, Peng X, Ma F, Yao Y, Liu X, Zhang Z, Yuan Z, Zhang L. A dynamic plasma membrane proteome analysis of alcohol-induced liver cirrhosis. Proteome Sci 2012; 10:39. [PMID: 22682408 PMCID: PMC3558348 DOI: 10.1186/1477-5956-10-39] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 05/25/2012] [Indexed: 12/26/2022] Open
Abstract
Alcohol-induced injury has become one of the major causes for liver cirrhosis. However, the molecular mechanisms of ethanol-induced injury are not fully understood. To this end, we performed a dynamic plasma membrane proteomic research on rat model. A rat model from hepatitis to liver cirrhosis was developed. Plasma membrane from liver tissue with liver fibrosis stage of 2 and 4 (S2 and S4) was purified by sucrose density gradient centrifugation. Its purification was verified by western blotting. Proteins from plasma membrane were separated by two-dimensional electrophoresis (2DE) and differentially expressed proteins were identified by tandem mass spectrometry. 16 consistent differentially expressed proteins from S2 to S4 were identified by mass spectrometry. The expression of differentially expressed proteins annexin A6 and annexin A3 were verified by western blotting, and annexin A3 was futher verified by immunohistochemistry. Our research suggests a possible mechanism by which ethanol alters protein expression to enhance the liver fibrosis progression. These differentially expressed proteins might be new drug targets for treating alcoholic liver cirrhosis.
Collapse
Affiliation(s)
- Xiaofang Jia
- Shanghai Public Health Clinical Center affiliated to Fudan University, Shanghai 201508, China
| | - Lin Yin
- Shanghai Public Health Clinical Center affiliated to Fudan University, Shanghai 201508, China
| | - Yanling Feng
- Shanghai Public Health Clinical Center affiliated to Fudan University, Shanghai 201508, China
| | - Xia Peng
- Shanghai Public Health Clinical Center affiliated to Fudan University, Shanghai 201508, China
| | - Fang Ma
- Shanghai Public Health Clinical Center affiliated to Fudan University, Shanghai 201508, China
| | - Yamin Yao
- Shanghai Public Health Clinical Center affiliated to Fudan University, Shanghai 201508, China
| | - Xiaoqian Liu
- Shanghai Public Health Clinical Center affiliated to Fudan University, Shanghai 201508, China
| | - Zhiyong Zhang
- Shanghai Public Health Clinical Center affiliated to Fudan University, Shanghai 201508, China
| | - Zhenghong Yuan
- Shanghai Public Health Clinical Center affiliated to Fudan University, Shanghai 201508, China
| | - Lijun Zhang
- Shanghai Public Health Clinical Center affiliated to Fudan University, Shanghai 201508, China.,Institute of Clinical Pharmacology, Pharmacogenetics Research Institute, Changsha, Hunan 410078, China
| |
Collapse
|
19
|
Hannivoort RA, Hernandez-Gea V, Friedman SL. Genomics and proteomics in liver fibrosis and cirrhosis. FIBROGENESIS & TISSUE REPAIR 2012; 5:1. [PMID: 22214245 PMCID: PMC3260086 DOI: 10.1186/1755-1536-5-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 01/03/2012] [Indexed: 12/15/2022]
Abstract
Genomics and proteomics have become increasingly important in biomedical science in the past decade, as they provide an opportunity for hypothesis-free experiments that can yield major insights not previously foreseen when scientific and clinical questions are based only on hypothesis-driven approaches. Use of these tools, therefore, opens new avenues for uncovering physiological and pathological pathways. Liver fibrosis is a complex disease provoked by a range of chronic injuries to the liver, among which are viral hepatitis, (non-) alcoholic steatohepatitis and autoimmune disorders. Some chronic liver patients will never develop fibrosis or cirrhosis, whereas others rapidly progress towards cirrhosis in a few years. This variety can be caused by disease-related factors (for example, viral genotype) or host-factors (genetic/epigenetic). It is vital to establish accurate tools to identify those patients at highest risk for disease severity or progression in order to determine who are in need of immediate therapies. Moreover, there is an urgent imperative to identify non-invasive markers that can accurately distinguish mild and intermediate stages of fibrosis. Ideally, biomarkers can be used to predict disease progression and treatment response, but these studies will take many years due to the requirement for lengthy follow-up periods to assess outcomes. Current genomic and proteomic research provides many candidate biomarkers, but independent validation of these biomarkers is lacking, and reproducibility is still a key concern. Thus, great opportunities and challenges lie ahead in the field of genomics and proteomics, which, if successful, could transform the diagnosis and treatment of chronic fibrosing liver diseases.
Collapse
Affiliation(s)
- Rebekka A Hannivoort
- Department of Medicine/Division of Liver Diseases, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | | | |
Collapse
|
20
|
Hua Y, Jia X, Sun M, Zheng L, Yin L, Zhang L, Cai Z. Plasma membrane proteomic analysis of human osteosarcoma and osteoblastic cells: revealing NDRG1 as a marker for osteosarcoma. Tumour Biol 2011; 32:1013-21. [PMID: 21706236 DOI: 10.1007/s13277-011-0203-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 06/10/2011] [Indexed: 01/07/2023] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumor in children and adolescents. To identify new biomarkers for early diagnosis of OS and novel therapeutic candidates, we carried out a plasma membrane proteomic study based on two-dimensional electrophoresis (2DE). The OS cell line MG-63 and the human osteoblastic cell line hFOB1.19 were adopted as the comparison model. We extracted plasma membrane by aqueous two-phase partition extraction. The proteins were separated through 2DE. We analyzed the differentially expressed proteins by Imagemaster software and then identified them by liquid chromatography-tandem mass spectrometry, and the location and function of differential proteins were searched through the Gene Ontology database. In total, 220 protein spots were separated by 2DE. Seven proteins with more than 2.0-folds of difference were successfully identified from 13 gel spots, with 6 up-regulated and 1 down-regulated. Gene Ontology analysis of the differentially expressed proteins indicated that these proteins were involved in seven kinds of functions including binding, structural, cell motility, receptor activity, electron carrier activity, NADH dehydrogenase (ubiquinone) activity, and transcription repressor activity. The up-regulation of NDRG1 was verified in osteosarcoma through Western blotting and by immunohistochemistry in paraffin-embedded tissues. The plasma membrane proteins identified in this study may provide new insights into osteosarcoma cancer biology and potential diagnostic and therapeutic biomarkers.
Collapse
Affiliation(s)
- Yingqi Hua
- Musculoskeletal Oncology Center, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | | | | | | | | | | | | |
Collapse
|