1
|
Wang L, Wei Z, Wu Y, Hu X, Zhou L, Zhao M, Sun A, Shao G, Yang W, Lin Q. Nuclear receptor coactivator 7 (NCOA7) protects cancer cells from oxidative damage through its ERbd domain. Cell Signal 2024; 124:111382. [PMID: 39243920 DOI: 10.1016/j.cellsig.2024.111382] [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: 04/20/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Oxidative stress causes damage to cancer cells and plays an important role in cancer therapy. Antagonizing oxidative stress is crucial for cancer cells to survive during the oxidation-based therapy. In this study, we defined the role of nuclear receptor co-activator 7 (NCOA7) in anti-oxidation in lung cancer cells and found that NCOA7 protects lung cancer A549 cells from the oxidative damage caused by hydrogen peroxide. Knockdown of NCOA7 in A549 cells significantly enhanced the hydrogen peroxide-caused inhibition of cell proliferation and migration, and markedly increased the damage effect of hydrogen peroxide on F-actin and focal adhesion structure, suggesting that NCOA7 protects F-actin and focal adhesion structure, thus the cell proliferation and migration, from oxidation-caused damage. Mechanistically, the anti-oxidation effect of NCOA7 is mediated by its nuclear receptor binding domain, the ERbd domain, suggesting that the anti-oxidation function of NCOA7 is dependent on its nuclear receptor co-activator activity. Our studies identified NCOA7 as an anti-oxidative protein through its nuclear receptor co-activator function and revealed the mechanism underlying the anti-oxidative effect of NCOA7 on cancer cell proliferation and migration.
Collapse
Affiliation(s)
- Lincui Wang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China; Department of Laboratory Medicine, the Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhixiao Wei
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yumeng Wu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiao Hu
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Liming Zhou
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Manhan Zhao
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Aiqin Sun
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Genbao Shao
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wannian Yang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Qiong Lin
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
2
|
Peng Z, Li Y, Xia S, Dai Q, Yin L, Chen M, Yang W, Shao G, Lin Q. Expression of nuclear receptor co‑activator 7 protein is associated with poor prognosis of breast cancer. Oncol Lett 2024; 27:278. [PMID: 38699661 PMCID: PMC11063752 DOI: 10.3892/ol.2024.14411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 03/25/2024] [Indexed: 05/05/2024] Open
Abstract
Nuclear receptor coactivator 7 (NCOA7) is an estrogen receptor binding protein. Its role in breast cancer progression has so far remained elusive. The present study aimed to determine the expression levels of NCOA7 in breast tumor samples and confirmed its potential utility as a breast cancer prognostic biomarker. The expression of NCOA7 was detected by immunohistochemical staining in 241 breast cancer tumor samples and 163 adjacent normal tissue samples. The association of NCOA7 expression with the clinicopathological characteristics and overall survival were statistically analyzed. Cell proliferation was determined by Cell Counting Kit-8 and colony-formation assays. Cell migration was detected using wound-healing and Transwell assays. NCOA7 was positively expressed in 44% of breast tumor tissues. The expression of NCOA7 was positively associated with tumor size (T-stage; P=0.005) and lymph node metastasis (N-stage; P=0.008). Additional statistical analysis indicated that the expression of NCOA7 was associated with patient age, tumor size and lymph node metastasis in patients with triple-negative breast cancer (TNBC) compared with that in patients with non-TNBC. The overall survival of patients with NCOA7-positive breast cancer was significantly lower than that of patients with NCOA7-negative breast cancer (P=0.006). Among the patients with lymph node metastasis, the overall survival was reversely associated with the expression of NCOA7 (P=0.042). Furthermore, knockdown of NCOA7 expression in breast cancer T47D and MCF7 cells significantly inhibited both cell proliferation and migration, suggesting that this protein may exert a role in driving breast cancer progression. Taken together, these results indicate that the expression of NCOA7 is associated with poor prognosis of breast cancer and suggest that this protein may be a driver for metastasis and a potential therapeutic target for advanced breast cancer.
Collapse
Affiliation(s)
- Ziluo Peng
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Yanlin Li
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Song Xia
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Qian Dai
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Liang Yin
- Department of Breast Surgery, Jiangsu University Affiliated People's Hospital, Zhenjiang, Jiangsu 212050, P.R. China
| | - Miao Chen
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
- Department of Pathology, Jiangsu University Affiliated People's Hospital, Zhenjiang, Jiangsu 212050, P.R. China
| | - Wannian Yang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Genbao Shao
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Qiong Lin
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
3
|
Guo J, Ke S, Chen Q, Zhou J, Guo J, Qiu T. NCOA7 Regulates Growth and Metastasis of Clear Cell Renal Cell Carcinoma via MAPK/ERK Signaling Pathway. Int J Mol Sci 2023; 24:11584. [PMID: 37511343 PMCID: PMC10380801 DOI: 10.3390/ijms241411584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
NCOA7 is a nuclear receptor coactivator that is downregulated in a variety of cancers. However, the expression and prognostic significance of NCOA7 in clear cell renal cell carcinoma (ccRCC) remain unknown. The expression of NCOA7 in ccRCC tissues was analyzed using bioinformatics analysis, Western blotting, and immunohistochemistry. Kaplan-Meier analysis, the receiver operating characteristic (ROC) curve, and clinicopathological correlation analysis were used to assess the predictive power of NCOA7. Overexpression function tests were conducted in cells and mouse models to clarify the function and mechanism of NCOA7 in inhibiting the progression of ccRCC. NCOA7 expression was downregulated in all three subtypes of renal cell carcinoma, and only had significant prognostic value for patients with ccRCC. NCOA7 overexpression inhibited the proliferation, invasion, and metastasis of ccRCC cells in vivo and in vitro. Mechanistically, NCOA7 inhibited the MAPK/ERK pathway to regulate epithelial-mesenchymal transformation (EMT) and apoptosis, thereby inhibiting the progression of ccRCC. NCOA7 inhibits tumor growth and metastasis of ccRCC through the MAPK/ERK pathway, thus indicating its potential as a prognostic marker and therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Jiayu Guo
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shuai Ke
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Qi Chen
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiangqiao Zhou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jia Guo
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tao Qiu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
4
|
Liu Y, Huang D, Li Z, Zhou L, Cen T, Wei B, Wei L, Wu H, Su L, Sooranna SR, Pan X, Huang Z. A plasma proteomic approach in patients with heart failure after acute myocardial infarction: insights into the pathogenesis and progression of the disease. Front Cardiovasc Med 2023; 10:1153625. [PMID: 37265567 PMCID: PMC10229768 DOI: 10.3389/fcvm.2023.1153625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 04/27/2023] [Indexed: 06/03/2023] Open
Abstract
Aims The pathogenesis of disease progression targets for patients with heart failure after acute myocardial infarction was investigated by using plasma proteomics. Methods The plasma proteomes of acute myocardial infarction patients with (MI-HF) and without (MI-WHF) heart failure were compared. Each group consisted of 10 patients who were matched for age and sex. The peptides were analyzed by 2-dimensional liquid chromatography coupled to tandem mass spectrometry in a high definition mode. Parallel reaction monitoring (PRM) verified the selected target proteins. Results We identified and quantified 2,589 and 2,222 proteins, respectively, and found 117 differentially expressed proteins (DEPs) (≥1.5-fold), when the MI-HF and MI-WHF groups were compared. Of these 51 and 66 were significantly up-regulated and down-regulated, respectively. The significant DEPs was subjected to protein-protein interaction network analysis which revealed a central role of the NF-κB signaling pathway in the MI-HF patients. PRM verified that MB, DIAPH1, VNN1, GOT2, SLC4A1, CRP, CKM, SOD3, F7, DLD, PGAM2, GOT1, UBA7 and HYOU1 were 14 proteins which were highly expressed in MI-HF patients. Conclusions These findings showed a group of proteins related to the NF-κB signaling pathway in the pathogenesis of patients with poor outcomes after experiencing MI-HF. These proteins may be useful candidate markers for the diagnosis of MI-HF as well as help to elucidate the pathophysiology of this major cause of mortality in older patients.
Collapse
Affiliation(s)
- Yan Liu
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Da Huang
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Zhile Li
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - LiuFang Zhou
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Tuan Cen
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Baomin Wei
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Liuqing Wei
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Hongying Wu
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - Liye Su
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Graduate School, Youjiang Medical University for Nationalities, Baise, China
| | - Suren R. Sooranna
- Department of Surgery and Cancer, Imperial College London, Chelsea and Westminster Hospital, London, United Kingdom
- Life Science and Clinical Research Center, Youjiang Medical University for Nationalities, Baise, China
| | - Xinshou Pan
- Department of Cardiology, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
| | - ZhaoHe Huang
- Graduate School, Youjiang Medical University for Nationalities, Baise, China
- Affiliated Southwest Hospital, Youjiang Medical University for Nationalities, Baise, China
| |
Collapse
|
5
|
Jo A, Bae JH, Yoon YJ, Chung TH, Lee EW, Kim YH, Joh HM, Chung JW. Plasma-activated medium induces ferroptosis by depleting FSP1 in human lung cancer cells. Cell Death Dis 2022; 13:212. [PMID: 35256587 PMCID: PMC8901787 DOI: 10.1038/s41419-022-04660-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/10/2021] [Accepted: 02/11/2022] [Indexed: 12/18/2022]
Abstract
Cold atmospheric plasma (CAP) that generates reactive oxygen species (ROS) has received considerable scientific attentions as a new type of anticancer. In particular, an indirect treatment method of inducing cancer cell death through plasma-activated medium (PAM), rather than direct plasma treatment has been well established. Although various cell death pathways such as apoptosis, necroptosis, and autophagy have been suggested to be involved in PAM-induced cell death, the involvement of ferroptosis, another type of cell death regulated by lipid ROS is largely unknown. This study reports, that PAM promotes cell death via ferroptosis in human lung cancer cells, and PAM increases intracellular and lipid ROS, thereby resulting in mitochondrial dysfunction. The treatment of cells with N-acetylcysteine, an ROS scavenging agent, or ferrostatin-1, a ferroptosis inhibitor, protects cells against PAM-induced cell death. Interestingly, ferroptosis suppressor protein 1 (FSP1) is downregulated upon PAM treatment. Furthermore, the treatment of cells with iFSP1, an inhibitor of FSP1, further enhances PAM-induced ferroptosis. Finally, this study demonstrates that PAM inhibits tumor growth in a xenograft model with an increase in 4-hydroxynoneal and PTGS2, a byproduct of lipid peroxidation, and a decrease in FSP1 expression. This study will provide new insights into the underlying mechanism and therapeutic strategies of PAM-mediated cancer treatment.
Collapse
Affiliation(s)
- Ara Jo
- Department of Biological Sciences, Dong-A University, Busan, 49315, Korea.,Department of Molecular Biology and Immunology, College of Medicine, Kosin University, Busan, Korea
| | - Jin Hee Bae
- Department of Materials Physics, Dong-A University, Busan, 49315, Korea
| | - Yu Jeong Yoon
- Department of Molecular Biology and Immunology, College of Medicine, Kosin University, Busan, Korea
| | - Tae Hun Chung
- Department of Materials Physics, Dong-A University, Busan, 49315, Korea
| | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Korea.,Department of Functional Genomics, University of Science and Technology (UST), Daejeon, 34141, Korea
| | - Young-Ho Kim
- Department of Molecular Biology and Immunology, College of Medicine, Kosin University, Busan, Korea
| | - Hea Min Joh
- Department of Materials Physics, Dong-A University, Busan, 49315, Korea.
| | - Jin Woong Chung
- Department of Biological Sciences, Dong-A University, Busan, 49315, Korea.
| |
Collapse
|
6
|
Integrated Proteomics Based on 2D Gel Electrophoresis and Mass Spectrometry with Validations: Identification of a Biomarker Compendium for Oral Submucous Fibrosis—An Indian Study. J Pers Med 2022; 12:jpm12020208. [PMID: 35207696 PMCID: PMC8878868 DOI: 10.3390/jpm12020208] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 12/13/2022] Open
Abstract
Oral Submucous Fibrosis (OSMF) is a chronic debilitating disease more frequently found in the South East Asian population. This disease poses a public health priority, as it is grouped under oral potentially malignant disorders, with malignant transformation rates of around 7 to 13%. Hence, early identification of high-risk OSMF patients is of the utmost importance to prevent malignant transformation. Proteomic expression profiling is a promising method for identifying differentially expressed proteins for disease prognosis and risk stratification in OSMF. In this study, overexpressed proteins in OSMF, OSMF transformed into oral squamous cell carcinoma (OSCC) and normal tissues were evaluated by proteomic analysis using two-dimensional electrophoresis (2DE) and mass spectrometry, which revealed 23 upregulated proteins. Validation was done using immunohistochemistry for three secretory proteins, namely 14-3-3ε (n = 130), carbonic anhydrase 1 (CA 1) (n = 125) and heat shock protein 70 (HSP 70) (n = 117), which showed significant overexpression in OSMF, OSCC compared to normal. The present study is the first of its kind in India to the best of our knowledge, assessing the altered expression of proteins in OSMF and OSMF which has undergone malignant transformation, obtaining a better knowledge of the molecular pathways involved in the disease progression. The current study shows that the biomarkers studied can be potentially useful for risk stratification of OSMF to OSCC serving as novel targets for therapeutic intervention. Clinical validation of the targets can further pave way for precision medicine to improve the quality of life in OSMF patients.
Collapse
|
7
|
Pillai J, Chincholkar T, Dixit R, Pandey M. A systematic review of proteomic biomarkers in oral squamous cell cancer. World J Surg Oncol 2021; 19:315. [PMID: 34711249 PMCID: PMC8555221 DOI: 10.1186/s12957-021-02423-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/06/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Head and neck squamous cell cancer (HNSCC) is the most common cancer associated with chewing tobacco, in the world. As this is divided in to sites and subsites, it does not make it to top 10 cancers. The most common subsite is the oral cancer. At the time of diagnosis, more than 50% of patients with oral squamous cell cancers (OSCC) had advanced disease, indicating the lack of availability of early detection and risk assessment biomarkers. The new protein biomarker development and discovery will aid in early diagnosis and treatment which lead to targeted treatment and ultimately a good prognosis. METHODS This systematic review was performed as per PRISMA guidelines. All relevant studies assessing characteristics of oral cancer and proteomics were considered for analysis. Only human studies published in English were included, and abstracts, incomplete articles, and cell line or animal studies were excluded. RESULTS A total of 308 articles were found, of which 112 were found to be relevant after exclusion. The present review focuses on techniques of cancer proteomics and discovery of biomarkers using these techniques. The signature of protein expression may be used to predict drug response and clinical course of disease and could be used to individualize therapy with such knowledge. CONCLUSIONS Prospective use of these markers in the clinical setting will enable early detection, prediction of response to treatment, improvement in treatment selection, and early detection of tumor recurrence for disease monitoring. However, most of these markers for OSCC are yet to be validated.
Collapse
Affiliation(s)
| | | | - Ruhi Dixit
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221 005, India
| | - Manoj Pandey
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221 005, India.
| |
Collapse
|
8
|
Singh AG, Roy S, Oza S, Singhavi H, Chatterjee K, Chaturvedi P. A contemporary narrative review to guide molecular epidemiology of oral submucous fibrosis. INTERNATIONAL JOURNAL OF MOLECULAR EPIDEMIOLOGY AND GENETICS 2021; 12:61-70. [PMID: 34552689 PMCID: PMC8449189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/09/2021] [Indexed: 06/13/2023]
Abstract
Oral submucous Fibrosis (OSMF) is a chronic disease that mainly affects the upper part of the aerodigestive tract. Areca nut and betel quid chewing has been established as the most significant causative factor for this condition. While OSMF is a predominantly Asian disease, the migrant populations from the region have taken the disease across the globe. Additionally, areca nut is now easily accessible in flavors and aggressively marketed. Many research activities have been undertaken for decades to understand the etiopathogenesis and risk factors of OSMF. Although OSMF is a slowly progressing disease, it has the potential to transform to an oral malignancy. This article is an attempt to review the literature and provide an update on its prevalence, etiopthogenesis and its diagnosis. We also highlight certain clinical, histopathological and molecular features that aid in the diagnosis and prognostication of OSMF, highlighting the importance of identifying the possibly high risk OSMF that is prone to malignant transformation. Using this information, future directions can be developed to include treatmentof OSMF through a dynamic gene-specific approach.
Collapse
|
9
|
Kerr AR, Lodi G. Management of Oral Potentially Malignant Disorders. Oral Dis 2021; 27:2008-2025. [PMID: 34324758 DOI: 10.1111/odi.13980] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/07/2021] [Accepted: 07/22/2021] [Indexed: 11/29/2022]
Abstract
Patients with oral potentially malignant disorders (OPMDs), including oral leukoplakia and erythroplakia, proliferative verrucous leukoplakia, oral submucous fibrosis, and oral lichen planus/lichenoid lesions can be challenging to manage. A small proportion will undergo cancer development and determining a patient's cancer risk is key to making management decisions. Yet, our understanding of the natural history of OPMDs has not been fully elucidated, and a precision approach based on the integration of numerous predictive markers has not been validated by prospective studies. Evidence-based health promotion by clinicians and healthcare systems is not embraced universally. Medical and surgical interventions evaluated by rigorous research measuring important endpoints, such as cancer development, mortality, or survival are difficult and expensive to run. Most of these studies employ non-ideal surrogate endpoints and have deep methodologic flaws. Diagnostic criteria for enrolling research subjects are not uniform, and patients with the highest risk for cancer development comprise small proportions of those enrolled. Few studies explore quality of life and patient preferences. It is time to rethink how we approach the management of these patients, across each OPMD, and considering the healthcare infrastructure and cost effectiveness. Global networks with well-characterized patient populations with OPMDs and well-designed interventional trials using validated outcome measures are needed.
Collapse
Affiliation(s)
- A Ross Kerr
- Department of Oral & Maxillofacial Pathology, Radiology & Medicine.,New York University College of Dentistry, New York, NY, USA
| | - Giovanni Lodi
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, Milano, Italia
| |
Collapse
|
10
|
Wang Y, Luo D, Yuan X, Luo Y, Cheng X, Gao Y, Xie X. Oxidative-protective effect of nuclear receptor coactivator 7 on arecoline-induced endothelial-to-mesenchymal transition. Oral Surg Oral Med Oral Pathol Oral Radiol 2020; 130:565-573. [PMID: 32988808 DOI: 10.1016/j.oooo.2020.08.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/02/2020] [Accepted: 08/13/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Overproduction of reactive oxygen species (ROS) has been implicated in inflammatory activities and tumorigenesis in oral submucous fibrosis (OSF). Nuclear receptor coactivator 7 (NCOA7) is capable of regulating cellular responses to ROS. The aim of this study was to investigate the expression of NCOA7 in endothelial cells and the role of NCOA7 in areca nut-induced endothelial-to-mesenchymal transition (EndMT). STUDY DESIGN Immunohistochemistry and immunofluorescence were used to detect the expression of NCOA7 in endothelia. Human umbilical vein endothelial cells (HUVECs) were treated with various dosages of arecoline (0, 5, 10, 20 μg/mL); then NCOA7 expression, the correlation of NCOA7 with EndMT, and the potential signaling were analyzed by using small interfering RNA (siRNA) transfection, reverse transcription polymerase chain reaction, Western blotting, and flow cytometry. RESULTS NCOA7 was significantly elevated in OSF tissues, as detected with immunohistochemistry and immunofluorescence. After arecoline treatment, NCOA7 expression and EndMT were induced in HUVECs. Transfection of HUVECs with si-NCOA7, which reduced 73% of NCOA7 expression, aggravated the arecoline-induced EndMT process. Inhibition of ROS markedly, but not completely, reverses this arecoline-induced EndMT in si-NCOA7 cells. CONCLUSIONS This study highlights NCOA7 as a potential target for therapeutic intervention to mediate EndMT via ROS species production.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Endodontics, Shenzhen Stomatology Hospital, Shenzhen, P. R. China
| | - Di Luo
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, P. R. China
| | - Xun Yuan
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, P. R. China
| | - Yin Luo
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, P. R. China
| | - Xiufeng Cheng
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, P. R. China
| | - Yijun Gao
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, P. R. China
| | - Xiaoyan Xie
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, P. R. China.
| |
Collapse
|
11
|
Shen YW, Shih YH, Fuh LJ, Shieh TM. Oral Submucous Fibrosis: A Review on Biomarkers, Pathogenic Mechanisms, and Treatments. Int J Mol Sci 2020; 21:ijms21197231. [PMID: 33008091 PMCID: PMC7582467 DOI: 10.3390/ijms21197231] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/24/2020] [Accepted: 09/26/2020] [Indexed: 02/07/2023] Open
Abstract
Oral submucous fibrosis (OSF) is a collagen deposition disorder that affects a patient’s oral function and quality of life. It may also potentially transform into malignancy. This review summarizes the risk factors, pathogenic mechanisms, and treatments of OSF based on clinical and bio-molecular evidence. Betel nut chewing is a major risk factor that causes OSF in Asia. However, no direct evidence of arecoline-induced carcinogenesis has been found in animal models. Despite identification of numerous biomarkers of OSF lesions and conducting trials with different drug combinations, clinicians still adopt conservative treatments that primarily focus on relieving the symptoms of OSF. Treatments focus on reducing inflammation and improving mouth opening to improve a patient’s quality of life. In conclusion, high-quality clinical studies are needed to aid clinicians in developing and applying molecular biomarkers as well as standard treatment guidelines.
Collapse
Affiliation(s)
- Yen-Wen Shen
- School of Dentistry, China Medical University, Taichung 40402, Taiwan;
- Department of Dentistry, China Medical University Hospital, Taichung City 404332, Taiwan
| | - Yin-Hwa Shih
- Department of Healthcare Administration, Asia University, Taichung 41354, Taiwan;
| | - Lih-Jyh Fuh
- School of Dentistry, China Medical University, Taichung 40402, Taiwan;
- Department of Dentistry, China Medical University Hospital, Taichung City 404332, Taiwan
- Correspondence: (L.-J.F.); (T.-M.S.); Tel.: +88-642-205-3366 (ext. 2312) (L.-J.F.); +88-642-205-3366 (ext. 7707) (T.-M.S.)
| | - Tzong-Ming Shieh
- School of Dentistry, China Medical University, Taichung 40402, Taiwan;
- Department of Dental Hygiene, China Medical University, Taichung 40402, Taiwan
- Correspondence: (L.-J.F.); (T.-M.S.); Tel.: +88-642-205-3366 (ext. 2312) (L.-J.F.); +88-642-205-3366 (ext. 7707) (T.-M.S.)
| |
Collapse
|
12
|
Gargaro M, Vacca C, Massari S, Scalisi G, Manni G, Mondanelli G, Mazza EMC, Bicciato S, Pallotta MT, Orabona C, Belladonna ML, Volpi C, Bianchi R, Matino D, Iacono A, Panfili E, Proietti E, Iamandii IM, Cecchetti V, Puccetti P, Tabarrini O, Fallarino F, Grohmann U. Engagement of Nuclear Coactivator 7 by 3-Hydroxyanthranilic Acid Enhances Activation of Aryl Hydrocarbon Receptor in Immunoregulatory Dendritic Cells. Front Immunol 2019; 10:1973. [PMID: 31481962 PMCID: PMC6710348 DOI: 10.3389/fimmu.2019.01973] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 08/05/2019] [Indexed: 12/18/2022] Open
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) catalyzes the first step in the kynurenine pathway of tryptophan (Trp) degradation that produces several biologically active Trp metabolites. L-kynurenine (Kyn), the first byproduct by IDO1, promotes immunoregulatory effects via activation of the Aryl hydrocarbon Receptor (AhR) in dendritic cells (DCs) and T lymphocytes. We here identified the nuclear coactivator 7 (NCOA7) as a molecular target of 3-hydroxyanthranilic acid (3-HAA), a Trp metabolite produced downstream of Kyn along the kynurenine pathway. In cells overexpressing NCOA7 and AhR, the presence of 3-HAA increased the association of the two molecules and enhanced Kyn-driven, AhR-dependent gene transcription. Physiologically, conventional (cDCs) but not plasmacytoid DCs or other immune cells expressed high levels of NCOA7. In cocultures of CD4+ T cells with cDCs, the co-addition of Kyn and 3-HAA significantly increased the induction of Foxp3+ regulatory T cells and the production of immunosuppressive transforming growth factor β in an NCOA7-dependent fashion. Thus, the co-presence of NCOA7 and the Trp metabolite 3-HAA can selectively enhance the activation of ubiquitary AhR in cDCs and consequent immunoregulatory effects. Because NCOA7 is often overexpressed and/or mutated in tumor microenvironments, our current data may provide evidence for a new immune check-point mechanism based on Trp metabolism and AhR.
Collapse
Affiliation(s)
- Marco Gargaro
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Carmine Vacca
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Serena Massari
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Giulia Scalisi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Giorgia Manni
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Giada Mondanelli
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Emilia M C Mazza
- Laboratory of Translational Immunology, Istituto Clinico Humanitas IRCCS, Rozzano, Italy
| | - Silvio Bicciato
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Maria T Pallotta
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Ciriana Orabona
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Maria L Belladonna
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Claudia Volpi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Roberta Bianchi
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Davide Matino
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Alberta Iacono
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Eleonora Panfili
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Elisa Proietti
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Violetta Cecchetti
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Paolo Puccetti
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Oriana Tabarrini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | | | - Ursula Grohmann
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|
13
|
Lu Z, Ding L, Ding H, Hao F, Pu Y, Wang Y, Chen S, Yang Y, Zhao X, Huang X, Zhang L, Wang Z, Hu Q, Ni Y. Tumor cell-derived TGF-β at tumor center independently predicts recurrence and poor survival in oral squamous cell carcinoma. J Oral Pathol Med 2019; 48:696-704. [PMID: 31141218 DOI: 10.1111/jop.12888] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 05/05/2019] [Accepted: 05/27/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Transforming growth factor-β (TGF-β) exerts its versatile function (oncogenic or tumor suppressive role) during the carcinogenesis in tumor microenvironment-dependent manner. Considering the tumor heterogeneity, spatial and temporal distribution of TGF-β in oral squamous cell carcinoma (OSCC) remained to be elucidated. METHODS Formalin-fixed, paraffin-embedded sections derived from 73 patients with OSCC were immunostained, revealing expression patterns of TGF-β, both at the regions of tumor center (TC) and invasive tumor front (ITF). RESULTS The TGF-β levels on tumor cells, fibroblast-like cells (FLCs), and tumor-infiltrating lymphocytes (TILs) were comparable and showed to be cell-type-independent manner. Although TC regions harbored less positive staining of TGF-β than ITF in tumor cells (TGF-βTumor cell ) (89.0% vs 98.3%; P = 0.037), FLCs (TGF-βFLC ) (86.3% vs 96.6%; P = 0.043), and TILs (TGF-βTIL ) (83.6% vs 94.8%; P = 0.044), respectively, TGF-β at TC regions, not at ITF, correlated to poor clinical outcomes. At TC regions, patients with high TGF-βTumor cell had high recurrence rate, and patients with high TGF-βTIL showed inferior worst pattern of invasion. Of note, high TGF-βTumor cell at TC predicted shorter overall survival time, recurrence-free survival, and disease-free survival in patients with OSCC, whereas high TGF-βTIL had no association with survival time. Cox regression analyses indicated that tumor cell-derived TGF-β at TC was an independent risk factor for survival outcome in patients with OSCC. CONCLUSIONS Tumor cell-derived TGF-β at TC regions, but not at ITF, could be a promising predictor for disease recurrence and poor prognosis of patients with OSCC.
Collapse
Affiliation(s)
- Zhanyi Lu
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Liang Ding
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Haoyue Ding
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Fengyao Hao
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yumei Pu
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yujia Wang
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Sheng Chen
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yan Yang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xingxing Zhao
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xiaofeng Huang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Lei Zhang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhiyong Wang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Qingang Hu
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yanhong Ni
- Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
14
|
Oral Submucous Fibrosis: A Review on Etiopathogenesis, Diagnosis, and Therapy. Int J Mol Sci 2019; 20:ijms20122940. [PMID: 31208114 PMCID: PMC6627879 DOI: 10.3390/ijms20122940] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 06/10/2019] [Accepted: 06/14/2019] [Indexed: 12/24/2022] Open
Abstract
Oral submucous fibrosis (OSF) is characterized by abnormal collagen deposition. It is a precancerous disorder and transforms into a malignant tumor in 1.5–15% of all cases. Symptoms include submucous fibrosis, ulceration, xerostomia, a burning sensation, and restricted mouth opening. All of these greatly interfere with patient quality of life. The present review introduces OSF from a molecular perspective and summarizes what is known about its underlying mechanisms, diagnostic biomarkers, and therapeutic interventions. In addition to the aggressive treatment of OSF, its prevention is also important. Future research should, therefore, focus on improving the oral health literacy of the patients susceptible to OSF.
Collapse
|
15
|
Thutkawkorapin J, Lindblom A, Tham E. Exome sequencing in 51 early onset non-familial CRC cases. Mol Genet Genomic Med 2019; 7:e605. [PMID: 30809968 PMCID: PMC6503031 DOI: 10.1002/mgg3.605] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 12/22/2018] [Accepted: 01/16/2019] [Indexed: 12/12/2022] Open
Abstract
Background Colorectal cancer (CRC) cases with an age of onset <40 years suggests a germline genetic cause. In total, 51 simplex cases were included to test the hypothesis of CRC as a mendelian trait caused by either heterozygous autosomal dominant or bi‐allelic autosomal recessive pathogenic variants. Methods The cohort was whole exome sequenced (WES) at 100× coverage. Both a dominant‐ and recessive model were used for searching predisposing genetic factors. In addition, we assayed recessive variants of potential moderate risk that were enriched in our young‐onset CRC cohort. Variants were filtered using a candidate cancer gene list or by selecting variants more likely to be pathogenic based on variant type (e.g., loss‐of‐function) or allele frequency. Results We identified one pathogenic variant in PTEN in a patient subsequently confirmed to have a hereditary hamartoma tumor syndrome (Cowden syndrome) and one patient with a pathogenic heterozygous variant in PMS2 that was originally not identified by WES due to low quality reads resulting from pseudogenes. In addition, we identified three heterozygous candidate missense variants in known cancer susceptibility genes (BMPR1A,BRIP1, and SRC), three truncating variants in possibly novel cancer genes (CLSPN,SEC24B, SSH2) and four candidate missense variants in ACACA, NR2C2, INPP4A, and DIDO1. We also identify five possible autosomal recessive candidate genes: ATP10B,PKHD1,UGGT2,MYH13,TFF3. Conclusion Two clear pathogenic variants were identified in patients that had not been identified clinically. Thus, the chance of detecting a hereditary cancer syndrome in patients with CRC at young age but without family history is 2/51 (4%) and therefore the clinical benefit of genetic testing in this patient group is low. Of note, using stringent filtering, we have identified a total of ten candidate heterozygous variants and five possibly biallelic autosomal recessive candidate genes that warrant further study.
Collapse
Affiliation(s)
| | - Annika Lindblom
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| | - Emma Tham
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Department of Clinical Genetics, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
16
|
Hoffmann F, Umbreit C, Krüger T, Pelzel D, Ernst G, Kniemeyer O, Guntinas-Lichius O, Berndt A, von Eggeling F. Identification of Proteomic Markers in Head and Neck Cancer Using MALDI-MS Imaging, LC-MS/MS, and Immunohistochemistry. Proteomics Clin Appl 2018; 13:e1700173. [PMID: 30411850 DOI: 10.1002/prca.201700173] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 10/29/2018] [Indexed: 12/12/2022]
Abstract
PURPOSE The heterogeneity of squamous cell carcinoma tissue greatly complicates diagnosis and individualized therapy. Therefore, characterizing the heterogeneity of tissue spatially and identifying appropriate biomarkers is crucial. MALDI-MS imaging (MSI) is capable of analyzing spatially resolved tissue biopsies on a molecular level. EXPERIMENTAL DESIGN MALDI-MSI is used on snap frozen and formalin-fixed and paraffin-embedded (FFPE) tissue samples from patients with head and neck cancer (HNC) to analyze m/z values localized in tumor and nontumor regions. Peptide identification is performed using LC-MS/MS and immunohistochemistry (IHC). RESULTS In both FFPE and frozen tissue specimens, eight characteristic masses of the tumor's epithelial region are found. Using LC-MS/MS, the peaks are identified as vimentin, keratin type II, nucleolin, heat shock protein 90, prelamin-A/C, junction plakoglobin, and PGAM1. Lastly, vimentin, nucleolin, and PGAM1 are verified with IHC. CONCLUSIONS AND CLINICAL RELEVANCE The combination of MALDI-MSI, LC-MS/MS, and subsequent IHC furnishes a tool suitable for characterizing the molecular heterogeneity of tissue. It is also suited for use in identifying new representative biomarkers to enable a more individualized therapy.
Collapse
Affiliation(s)
- Franziska Hoffmann
- Department of Otorhinolaryngology, Jena University Hospital, Jena, Germany
| | - Claudia Umbreit
- Department of Otorhinolaryngology, Jena University Hospital, Jena, Germany.,Institute of Forensic Medicine, Section Pathology, Jena University Hospital, Jena, Germany
| | - Thomas Krüger
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Daniela Pelzel
- Department of Otorhinolaryngology, Jena University Hospital, Jena, Germany
| | - Günther Ernst
- Department of Otorhinolaryngology, Jena University Hospital, Jena, Germany
| | - Olaf Kniemeyer
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | | | - Alexander Berndt
- Institute of Forensic Medicine, Section Pathology, Jena University Hospital, Jena, Germany
| | - Ferdinand von Eggeling
- Department of Otorhinolaryngology, Jena University Hospital, Jena, Germany.,Institute of Physical Chemistry, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
17
|
Jaegger CF, Negrão F, Assis DM, Belaz KRA, Angolini CFF, Fernandes AMAP, Santos VG, Pimentel A, Abánades DR, Giorgio S, Eberlin MN, Rocha DFO. MALDI MS imaging investigation of the host response to visceral leishmaniasis. MOLECULAR BIOSYSTEMS 2018; 13:1946-1953. [PMID: 28758666 DOI: 10.1039/c7mb00306d] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Mass spectrometry imaging (MSI) of animal tissues has become an important tool for in situ molecular analyses and biomarker studies in several clinical areas, but there are few applications in parasitological studies. Leishmaniasis is a neglected tropical disease, and experimental mouse models have been essential to evaluate pathological and immunological processes and to develop diagnostic methods. Herein we have employed MALDI MSI to examine peptides and low molecular weight proteins (2 to 20 kDa) differentially expressed in the liver during visceral leishmaniasis in mice models. We analyzed liver sections of Balb/c mice infected with Leishmania infantum using the SCiLS Lab software for statistical analysis, which facilitated data interpretation and thus highlighted several key proteins and/or peptides. We proposed a decision tree classification for visceral leishmaniasis with distinct phases of the disease, which are named here as healthy, acute infection and chronic infection. Among others, the ion of m/z 4963 was the most important to identify acute infection and was tentatively identified as Thymosin β4. This peptide was previously established as a recovery factor in the human liver and might participate in the response of mice to Leishmania infection. This preliminary investigation shows the potential of MALDI MSI to complement classical compound selective imaging techniques and to explore new features not yet recognized by these approaches.
Collapse
Affiliation(s)
- C F Jaegger
- ThoMSon Mass Spectrometry Laboratory, University of Campinas - UNICAMP, Campinas, SP, Brazil.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Yang H, Ji W, Guan M, Li S, Zhang Y, Zhao Z, Mao L. Organic washes of tissue sections for comprehensive analysis of small molecule metabolites by MALDI MS imaging of rat brain following status epilepticus. Metabolomics 2018; 14:50. [PMID: 30830331 DOI: 10.1007/s11306-018-1348-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 03/09/2018] [Indexed: 12/19/2022]
Abstract
INTRODUCTION In-situ detection and in particular comprehensive analysis of small molecule metabolites (SMMs, m/z < 500) using matrix-assisted laser desorption ionization mass spectrometry imaging (MALDI MSI) remain a challenge, mainly due to ion suppression effects from more abundant molecules in tissue section like lipids. OBJECTIVE A strategy based on organic washes to remove most ionization-suppressing lipids from tissue section was firstly explored for improved analysis of SMMs by MALDI MSI. METHODS The tissue sections after rinse with different organic solvents were analyzed by MALDI MSI, and the results were compared for the optimized washing conditions. RESULTS The rinse with chloroform for 15 s at - 20 °C significantly removed most glycerophospholipids and glycerolipids from tissue section. Consequentially, ATP-related energy metabolites, amino acids and derivatives, glucose derivatives, glycolysis pathway metabolites and other SMMs were able to be well-visualized with enhanced ion intensity and good reproducibility. The organic washes-based MALDI MSI was applied to the metabolic pathway analysis in rat brain following status epilepticus (SE) model, which was, as far as we know, the first report about in-situ detection of a broad range of metabolites in the model of SE by MALDI MSI technique. The alterations of cyclic adenosine monophosphate (cyclic AMP), inosine, glutamine, glutathione, taurine and spermine during SE were observed. CONCLUSION A simple organic washing protocol enables comprehensive analysis of tissue SMMs in MALDI MSI by removing ionization-suppressing lipids. The application in the SE model indicates that MALDI MSI analysis potentially provides new insight for understanding the disease mechanism.
Collapse
Affiliation(s)
- Hui Yang
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Beijing Mass Spectrum Center, Institute of Chemistry Chinese Academy of Sciences, Beijing, China
- Graduate School, University of Chinese Academy of Sciences, Beijing, China
| | - Wenliang Ji
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Beijing Mass Spectrum Center, Institute of Chemistry Chinese Academy of Sciences, Beijing, China
| | - Ming Guan
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Beijing Mass Spectrum Center, Institute of Chemistry Chinese Academy of Sciences, Beijing, China
- Graduate School, University of Chinese Academy of Sciences, Beijing, China
| | - Shilei Li
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Beijing Mass Spectrum Center, Institute of Chemistry Chinese Academy of Sciences, Beijing, China
- Graduate School, University of Chinese Academy of Sciences, Beijing, China
| | - Yangyang Zhang
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Beijing Mass Spectrum Center, Institute of Chemistry Chinese Academy of Sciences, Beijing, China
| | - Zhenwen Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Beijing Mass Spectrum Center, Institute of Chemistry Chinese Academy of Sciences, Beijing, China.
- Graduate School, University of Chinese Academy of Sciences, Beijing, China.
| | - Lanqun Mao
- Beijing National Laboratory for Molecular Sciences, CAS Research/Education Center for Excellence in Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Beijing Mass Spectrum Center, Institute of Chemistry Chinese Academy of Sciences, Beijing, China.
- Graduate School, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
19
|
Zhang X, Sun L. Anaphylatoxin C3a: A potential biomarker for esophageal cancer diagnosis. Mol Clin Oncol 2017; 8:315-319. [PMID: 29435296 DOI: 10.3892/mco.2017.1524] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 11/28/2017] [Indexed: 12/30/2022] Open
Abstract
Esophageal carcinoma is a common malignancy worldwide, with a low 5-year survival rate. As the majority of cases are diagnosed at an advanced stage, there is an urgent need for an effective biomarker for early diagnosis of esophageal cancer patients. Surface-enhanced laser desorption ionization time-of-flight mass spectrometry (SELDI-TOF-MS) was applied to detect the serum protein expression in esophageal cancer patients using ProteinChip software, and the results were analyzed and screened using Biomarker Patterns and SPSS16.0 software. The ELISA method was conducted to determine the concentration of anaphylatoxin C3a, which is one of the complement proteins, in the serum of esophageal cancer patients and non-esophageal cancer participants. A total of 144 effective differential expression protein peaks in the window of 1-10 kDa were obtained (P<0.05). M/Z 8,926.478 (P<10-6) protein peak was employed as the diagnostic biomarker for esophageal carcinoma. This established diagnostic biomarker has a sensitivity of 95% (19/20) and an accuracy of 100% (19/19) for positive prediction. The results suggested that anaphylatoxin C3a may be a promising biomarker in the diagnosis of esophageal carcinoma, and may play a key role in promoting esophageal carcinogenesis.
Collapse
Affiliation(s)
- Xu Zhang
- School of Pharmacy, Yancheng Teachers University, Yancheng, Jiangsu 224002, P.R. China
| | - Lingzhi Sun
- School of Pharmacy, Yancheng Teachers University, Yancheng, Jiangsu 224002, P.R. China
| |
Collapse
|
20
|
Recent advances in sample pre-treatment for emerging methods in proteomic analysis. Talanta 2017; 174:738-751. [DOI: 10.1016/j.talanta.2017.06.056] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 06/14/2017] [Accepted: 06/19/2017] [Indexed: 12/21/2022]
|
21
|
Biomarker MicroRNAs for Diagnosis of Oral Squamous Cell Carcinoma Identified Based on Gene Expression Data and MicroRNA-mRNA Network Analysis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2017; 2017:9803018. [PMID: 29098014 PMCID: PMC5623795 DOI: 10.1155/2017/9803018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/01/2017] [Accepted: 08/06/2017] [Indexed: 12/26/2022]
Abstract
Oral squamous cell carcinoma is one of the most malignant tumors with high mortality rate worldwide. Biomarker discovery is critical for early diagnosis and precision treatment of this disease. MicroRNAs are small noncoding RNA molecules which often regulate essential biological processes and are good candidates for biomarkers. By integrative analysis of both the cancer-associated gene expression data and microRNA-mRNA network, miR-148b-3p, miR-629-3p, miR-27a-3p, and miR-142-3p were screened as novel diagnostic biomarkers for oral squamous cell carcinoma based on their unique regulatory abilities in the network structure of the conditional microRNA-mRNA network and their important functions. These findings were confirmed by literature verification and functional enrichment analysis. Future experimental validation is expected for the further investigation of their molecular mechanisms.
Collapse
|
22
|
Global Proteomics-based Identification and Validation of Thymosin Beta-4 X-Linked as a Prognostic Marker for Head and Neck Squamous Cell Carcinoma. Sci Rep 2017; 7:9031. [PMID: 28831179 PMCID: PMC5567379 DOI: 10.1038/s41598-017-09539-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 07/26/2017] [Indexed: 12/14/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) represents a major health concern worldwide. We applied the matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) to analyze paired normal (N) and tumor (T) samples from head and neck squamous cell carcinoma as well as liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis in HNSCC cell lines to identify tumor-associated biomarkers. Our results showed a number of proteins found to be over-expressed in HNSCC. We identified thymosin beta-4 X-linked (TMSB4X) is one of the most significant candidate biomarkers. Higher TMSB4X expression in the tumor was found by N/T-paired HNSCC samples at both RNA and protein level. Overexpression of TMSB4X was found significantly associated with poor prognosis of overall survival (OS, P = 0.006) and recurrence-free survival (RFS, P = 0.013) in HNSCC patients. Silencing of TMSB4X expression in HNSCC cell line reduced the proliferation and invasion ability in vitro, as well as inhibited the cervical lymph node metastasis in vivo. Altogether, our global proteomics analysis identified that TMSB4X is a newly discovered biomarker in HNSCC whose functions resulted in enhanced proliferation and metastasis in vitro and in vivo. TMSB4X may be a potential therapeutic target for treating HNSCC patients.
Collapse
|
23
|
Wang J, Jin X, Liu J, Zhao K, Xu H, Wen J, Jiang L, Zeng X, Li J, Chen Q. The prognostic value of B7-H6 protein expression in human oral squamous cell carcinoma. J Oral Pathol Med 2017; 46:766-772. [PMID: 28437013 DOI: 10.1111/jop.12586] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2017] [Indexed: 02/05/2023]
Affiliation(s)
- Jiongke Wang
- State Key Laboratory of Oral Diseases; National Clinical Research Center for Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Xin Jin
- Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences; College of Stomatology; Chongqing Medical University; Chongqing China
| | - Jiajia Liu
- State Key Laboratory of Oral Diseases; National Clinical Research Center for Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Kui Zhao
- State Key Laboratory of Oral Diseases; National Clinical Research Center for Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Hao Xu
- State Key Laboratory of Oral Diseases; National Clinical Research Center for Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Jing Wen
- State Key Laboratory of Oral Diseases; National Clinical Research Center for Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Lu Jiang
- State Key Laboratory of Oral Diseases; National Clinical Research Center for Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Xin Zeng
- State Key Laboratory of Oral Diseases; National Clinical Research Center for Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Jing Li
- State Key Laboratory of Oral Diseases; National Clinical Research Center for Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases; National Clinical Research Center for Oral Diseases; West China Hospital of Stomatology; Sichuan University; Chengdu China
| |
Collapse
|
24
|
Finelli MJ, Oliver PL. TLDc proteins: new players in the oxidative stress response and neurological disease. Mamm Genome 2017; 28:395-406. [PMID: 28707022 PMCID: PMC5614904 DOI: 10.1007/s00335-017-9706-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 07/03/2017] [Indexed: 12/14/2022]
Abstract
Oxidative stress (OS) arises from an imbalance in the cellular redox state, which can lead to intracellular damage and ultimately cell death. OS occurs as a result of normal ageing, but it is also implicated as a common etiological factor in neurological disease; thus identifying novel proteins that modulate the OS response may facilitate the design of new therapeutic approaches applicable to many disorders. In this review, we describe the recent progress that has been made using a range of genetic approaches to understand a family of proteins that share the highly conserved TLDc domain. We highlight their shared ability to prevent OS-related cell death and their unique functional characteristics, as well as discussing their potential application as new neuroprotective factors. Furthermore, with an increasing number of pathogenic mutations leading to epilepsy and hearing loss being discovered in the TLDc protein TBC1D24, understanding the function of this family has important implications for a range of inherited neurological diseases.
Collapse
Affiliation(s)
- Mattéa J Finelli
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK
| | - Peter L Oliver
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford, OX1 3PT, UK.
| |
Collapse
|
25
|
Negrão F, de O. Rocha DF, Jaeeger CF, Rocha FJS, Eberlin MN, Giorgio S. Murine cutaneous leishmaniasis investigated by MALDI mass spectrometry imaging. MOLECULAR BIOSYSTEMS 2017; 13:2036-2043. [DOI: 10.1039/c7mb00411g] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The MALDI-IMS technique was applied to screen for peptides and low molecular weight proteins to unveil potential biomarkers for cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Fernanda Negrão
- ThoMSon Mass Spectrometry Laboratory
- University of Campinas – UNICAMP
- Campinas-SP
- Brazil
- Department of Animal Biology
| | | | - Caroline F. Jaeeger
- ThoMSon Mass Spectrometry Laboratory
- University of Campinas – UNICAMP
- Campinas-SP
- Brazil
| | - Francisca J. S. Rocha
- Department of Tropical Medicine
- Federal University of Pernambuco – UFPE
- Recife-PE
- Brazil
| | - Marcos N. Eberlin
- ThoMSon Mass Spectrometry Laboratory
- University of Campinas – UNICAMP
- Campinas-SP
- Brazil
| | - Selma Giorgio
- Department of Animal Biology
- Biology Institute
- University of Campinas UNICAMP
- Campinas-SP
- Brazil
| |
Collapse
|