1
|
Lin Y, Xu G, Li L, Xiang J, Zhai L. Incretin-based drugs decrease the incidence of prostate cancer in type 2 diabetics: A pooling-up analysis. Medicine (Baltimore) 2024; 103:e38018. [PMID: 38758855 PMCID: PMC11098233 DOI: 10.1097/md.0000000000038018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 04/05/2024] [Indexed: 05/19/2024] Open
Abstract
Incretin-based drugs, a class of Antidiabetic medications (ADMs) used in the treatment of type 2 diabetes, may affect the incidence of prostate cancer (PCa). But real-world evidence for this possible effect is lacking. Therefore, the aim of this study is to assess the effect of incretin-based drugs on the incidence of PCa, including glucagon-like peptide-1 (GLP-1) receptor agonists and dipeptidyl peptidase-4 (DPP-4) inhibitors. We searched PubMed, Embase, and Cochrane Library databases for eligible studies through September 2023. Two independent reviewers performed screening and data extraction. We used the Cochrane Handbook for Systematic Reviews and the Newcastle-Ottawa Scale (NOS) to assess the quality of included randomized controlled trials (RCTs) and cohort studies. We did a meta-analysis of available trial data to calculate overall risk ratios (RRs) for PCa. A total of 1238 articles were identified in our search. After screening for eligibility, 7 high-quality studies met the criteria for meta-analysis, including 2 RCTs and 5 cohort studies, with a total of 1165,738 patients. Compared with the control group, we found that incretin-based drugs reduced the relative risk of PCa by 35% (95% confidence interval (CI), 0.17-0.49; P = .0006). In subgroup analysis, the RR values for GLP-1 receptor agonists and DPP-4 inhibitors were 62% (95% CI, 0.45-0.85; P = .003) and 72% (95% CI, 0.46-1.12; P = .14), respectively. Incretin-based drugs are associated with lower incidence of prostate cancer and may have a preventive effect on prostate cancer in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Yuxiang Lin
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guangyong Xu
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liangyu Li
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jingyi Xiang
- Department of Infectious Diseases, Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lingyun Zhai
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
2
|
Kotnala S, Dhasmana A, Dhasmana S, Haque S, Yallapu MM, Tripathi MK, Jaggi M, Chauhan SC. A Systems Biology Approach Unveils a Critical Role of DPP4 in Upper Gastrointestinal Cancer Patient Outcomes. J Environ Pathol Toxicol Oncol 2024; 43:43-55. [PMID: 38505912 PMCID: PMC11419273 DOI: 10.1615/jenvironpatholtoxicoloncol.2023048056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024] Open
Abstract
Gastrointestinal (GI) cancers comprise of cancers that affect the digestive system and its accessory organs. The late detection and poor prognosis of GI cancer emphasizes the importance of identifying reliable and precise biomarkers for early diagnosis and prediction of prognosis. The membrane-bound glycoprotein dipeptidyl-peptidase 4 (DPP4), also known as CD26, is ubiquitously expressed and has a wide spectrum of biological roles. The role of DPP4/CD26 in tumor progression in different types of cancers remains elusive. However, the link between DPP4 and tumor-infiltrating cells, as well as its prognostic significance in malignancies, still require further investigation. This study was intended to elucidate the correlation of DPP4 expression and survival along with prognosis, followed by its associated enriched molecular pathways and immune cell marker levels in upper GI cancers. Results demonstrated a strong correlation between increased DPP4 expression and a worse prognosis in esophageal and gastric cancer and the co-expressed common genes with DPP4 were associated with crucial molecular pathways involved in tumorigenesis. Additionally, DPP4 was shown to be significantly linked to several immune infiltrating cell marker genes, including Macrophages (M1, M2 and Tumor Associated Macrophages), neutrophils, Treg, T-cell exhaustion, Th1 and Th2. Overall, our findings suggest that DPP4 may serve as a substantial prognostic biomarker, a possible therapeutic target, as well as it can play a critical role in the regulation of immune cell invasion in patients with gastroesophageal (esophageal, gastroesophageal junction and gastric) cancer. KEY WORDS: DPP4, integrated analysis, GI cancer, gastroesophageal cancer, gastroesophageal junction, prognosis.
Collapse
Affiliation(s)
- Sudhir Kotnala
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- Department of Biosciences and Cancer Research Institute, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, India
| | - Swati Dhasmana
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Murali M. Yallapu
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Manish K. Tripathi
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Subhash C. Chauhan
- Department of Immunology and Microbiology, School of Medicine, The University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| |
Collapse
|
3
|
Wen Y, Lin C, Hsiao C, Wang S, Huang H, Lin Y, Ho K, Chang L, Yang S, Chien M. Genetic variants of dipeptidyl peptidase IV are linked to the clinicopathologic development of prostate cancer. J Cell Mol Med 2023; 27:2507-2516. [PMID: 37533175 PMCID: PMC10468658 DOI: 10.1111/jcmm.17845] [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: 06/04/2023] [Revised: 06/27/2023] [Accepted: 06/30/2023] [Indexed: 08/04/2023] Open
Abstract
CD26/dipeptidyl peptidase IV (DPP4) is a multifunctional cell-surface glycoprotein widely found in many cell types, and a soluble form is present in body fluids. There is longstanding evidence indicating a tumour-promoting or -suppressive role of DPP4 in different cancer types. However, studies focusing on the impacts of genetic variants of DPP4 on cancers are very rare. Herein, we conducted a case-control study to evaluate whether single-nucleotide polymorphisms (SNPs) of DPP4 were associated with the risk or clinicopathologic development of prostate cancer (PCa). We genotyped four loci of DPP4 SNPs, including rs7608798 (A/G), rs3788979 (C/T), rs2268889 (T/C) and rs6741949 (G/C), using a TaqMan allelic discrimination assay in 704 PCa patients and 704 healthy controls. Our results showed that PCa patients with the DPP4 rs7608798 AG+GG genotype or rs2268889 TC+CC genotype had a higher risk of developing an advanced clinical primary tumour (cT) stage (adjusted odds ratio (AOR): 1.680, 95% confidence interval (CI): 1.062-2.659, p = 0.025; AOR: 1.693, 95% CI: 1.092-2.624, p = 0.018). Additionally, in The Cancer Genome Atlas (TCGA) database, we observed that lower DPP4 expression levels were correlated with higher Gleason scores, advanced cT and pathological stages, tumour metastasis, and shorter progression-free survival rates in PCa patients. Furthermore, overexpression of DPP4 suppressed migration/invasion of metastatic PC3 PCa cells. Our findings suggest that DPP4 levels may affect the progression of PCa, and the DPP4 rs7608798 and rs2268889 SNPs are associated with the clinicopathologic development of PCa in a Taiwanese population.
Collapse
Affiliation(s)
- Yu‐Ching Wen
- Department of Urology, School of MedicineCollege of Medicine and TMU Research Center of Urology and Kidney (TMU‐RCUK), Taipei Medical UniversityTaipeiTaiwan
- Department of UrologyWan Fang Hospital, Taipei Medical UniversityTaipeiTaiwan
| | - Chia‐Yen Lin
- Division of Urology, Department of SurgeryTaichung Veterans General HospitalTaichungTaiwan
- School of MedicineChung Shan Medical UniversityTaichungTaiwan
- School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Chi‐Hao Hsiao
- Department of Urology, School of MedicineCollege of Medicine and TMU Research Center of Urology and Kidney (TMU‐RCUK), Taipei Medical UniversityTaipeiTaiwan
- Department of UrologyWan Fang Hospital, Taipei Medical UniversityTaipeiTaiwan
| | - Shian‐Shiang Wang
- Division of Urology, Department of SurgeryTaichung Veterans General HospitalTaichungTaiwan
- School of MedicineChung Shan Medical UniversityTaichungTaiwan
- Department of Applied ChemistryNational Chi Nan UniversityNantouTaiwan
| | - Hsiang‐Ching Huang
- Graduate Institute of Medical SciencesCollege of Medicine, Taipei Medical UniversityTaipeiTaiwan
| | - Yung‐Wei Lin
- Department of Urology, School of MedicineCollege of Medicine and TMU Research Center of Urology and Kidney (TMU‐RCUK), Taipei Medical UniversityTaipeiTaiwan
- Department of UrologyWan Fang Hospital, Taipei Medical UniversityTaipeiTaiwan
- International Master/PhD Program in MedicineCollege of Medicine, Taipei Medical UniversityTaipeiTaiwan
| | - Kuo‐Hao Ho
- Graduate Institute of Medical SciencesCollege of Medicine, Taipei Medical UniversityTaipeiTaiwan
| | - Lun‐Ching Chang
- Department of Mathematical SciencesFlorida Atlantic UniversityBoca RatonFloridaUSA
| | - Shun‐Fa Yang
- Institute of MedicineChung Shan Medical UniversityTaichungTaiwan
- Department of Medical ResearchChung Shan Medical University HospitalTaichungTaiwan
| | - Ming‐Hsien Chien
- International Master/PhD Program in MedicineCollege of Medicine, Taipei Medical UniversityTaipeiTaiwan
- Pulmonary Research Center, Wan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
- Traditional Herbal Medicine Research CenterTaipei Medical University HospitalTaipeiTaiwan
- TMU Research Center of Cancer Translational MedicineTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
4
|
Díaz-Gómez JL, López-Castillo LM, Garcia-Lara S, Castorena-Torres F, Winkler R, Wielsch N, Aguilar O. Novel α-zein peptide fractions with in vitro cytotoxic activity against hepatocarcinoma. FOOD AND BIOPRODUCTS PROCESSING 2022. [DOI: 10.1016/j.fbp.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
5
|
Does DPP-IV Inhibition Offer New Avenues for Therapeutic Intervention in Malignant Disease? Cancers (Basel) 2022; 14:cancers14092072. [PMID: 35565202 PMCID: PMC9103952 DOI: 10.3390/cancers14092072] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/16/2022] [Accepted: 04/18/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary There is growing interest in identifying the effects of antidiabetic agents on cancer risk, progression, and anti-cancer treatment due to the long-term use of these medications and the inherently increased risk of malignancies in diabetic patients. Tumor development and progression are affected by multiple mediators in the tumor microenvironment, several of which may be proteolytically modified by the multifunctional protease dipeptidyl peptidase-IV (DPP-IV, CD26). Currently, low-molecular-weight DPP-IV inhibitors (gliptins) are used in patients with type 2 diabetes based on the observation that DPP-IV inhibition enhances insulin secretion by increasing the bioavailability of incretins. However, the DPP-IV-mediated cleavage of other biopeptides and chemokines is also prevented by gliptins. The potential utility of gliptins in other areas of medicine, including cancer, is therefore being evaluated. Here, we critically review the existing evidence on the role of DPP-IV inhibitors in cancer pathogenesis, their potential to be used in anti-cancer treatment, and the possible perils associated with this approach. Abstract Dipeptidyl peptidase IV (DPP-IV, CD26) is frequently dysregulated in cancer and plays an important role in regulating multiple bioactive peptides with the potential to influence cancer progression and the recruitment of immune cells. Therefore, it represents a potential contributing factor to cancer pathogenesis and an attractive therapeutic target. Specific DPP-IV inhibitors (gliptins) are currently used in patients with type 2 diabetes mellitus to promote insulin secretion by prolonging the activity of the incretins glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP). Nevertheless, the modulation of the bioavailability and function of other DPP-IV substrates, including chemokines, raises the possibility that the use of these orally administered drugs with favorable side-effect profiles might be extended beyond the treatment of hyperglycemia. In this review, we critically examine the possible utilization of DPP-IV inhibition in cancer prevention and various aspects of cancer treatment and discuss the potential perils associated with the inhibition of DPP-IV in cancer. The current literature is summarized regarding the possible chemopreventive and cytotoxic effects of gliptins and their potential utility in modulating the anti-tumor immune response, enhancing hematopoietic stem cell transplantation, preventing acute graft-versus-host disease, and alleviating the side-effects of conventional anti-tumor treatments.
Collapse
|
6
|
Incretin-Based Drugs and the Incidence of Prostate Cancer Among Patients with Type 2 Diabetes. Epidemiology 2022; 33:563-571. [PMID: 35394977 DOI: 10.1097/ede.0000000000001486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND There is some evidence that glucagon-like peptide 1 (GLP-1) receptor agonists and dipeptidyl peptidase-4 (DPP-4) inhibitors have chemopreventive effects on prostate cancer cells, but real-world evidence for this possible effect is lacking. Thus, the objective of this study was to estimate whether use of GLP-1 receptor agonists and DPP-4 inhibitors, separately, is associated with a decreased risk of prostate cancer among patients with type 2 diabetes. METHODS We assembled two new-user, active comparator cohorts using the United Kingdom Clinical Practice Research Datalink (2007 to 2019). The first cohort included 5063 initiators of GLP-1 receptor agonists and 112,955 of sulfonylureas. The second cohort included 53,529 initiators of DPP-4 inhibitors and 114,417 of sulfonylureas. We fit Cox proportional hazards models to estimate adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) for prostate cancer. We weighted the models using propensity score fine stratification, which considered over 50 potential confounders. RESULTS GLP-1 receptor agonists were associated with a decreased risk of prostate cancer when compared with sulfonylureas (incidence rates: 156.4 vs. 232.0 per 100,000 person-years, respectively; HR: 0.65, 95% CI: 0.43, 0.99). DPP-4 inhibitors were also associated with a decreased risk of prostate cancer when compared with sulfonylureas (incidence rates: 316.2 vs. 350.5 events per 100,000 person-years, respectively; HR: 0.90, CI: 0.81, 1.00). CONCLUSIONS The results of this study are consistent with the hypothesis that the use of GLP-1 receptor agonists and DPP-4 inhibitors, separately, may decrease the risk of prostate cancer when compared with the use of sulfonylureas.
Collapse
|
7
|
Huang J, Liu X, Wei Y, Li X, Gao S, Dong L, Rao X, Zhong J. Emerging Role of Dipeptidyl Peptidase-4 in Autoimmune Disease. Front Immunol 2022; 13:830863. [PMID: 35309368 PMCID: PMC8931313 DOI: 10.3389/fimmu.2022.830863] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/14/2022] [Indexed: 12/12/2022] Open
Abstract
Dipeptidyl-peptidase IV (DPP4), originally identified as an aminopeptidase in 1960s, is an ubiquitously expressed protease presented as either a membrane-bound or soluble form. DPP4 cleaves dipeptide off from the N-terminal of its substrates, altering the bioactivity of its substrates. Subsequent studies reveal that DPP4 is also involved in various cellular processes by directly binding to a number of ligands, including adenosine deaminase, CD45, fibronectin, plasminogen, and caveolin-1. In recent years, many novel functions of DPP4, such as promoting fibrosis and mediating virus entry, have been discovered. Due to its implication in fibrotic response and immunoregulation, increasing studies are focusing on the potential role of DPP4 in inflammatory disorders. As a moonlighting protein, DPP4 possesses multiple functions in different types of cells, including both enzymatic and non-enzymatic functions. However, most of the review articles on the role of DPP4 in autoimmune disease were focused on the association between DPP4 enzymatic inhibitors and the risk of autoimmune disease. An updated comprehensive summary of DPP4’s immunoregulatory actions including both enzymatic dependent and independent functions is needed. In this article, we will review the recent advances of DPP4 in immune regulation and autoimmune rheumatic disease.
Collapse
Affiliation(s)
- Jie Huang
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Xinxin Liu
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Yingying Wei
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Xinlu Li
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Shupei Gao
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Lingli Dong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jixin Zhong, ; Xiaoquan Rao, ; Lingli Dong,
| | - Xiaoquan Rao
- Department of Cardiovascular Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jixin Zhong, ; Xiaoquan Rao, ; Lingli Dong,
| | - Jixin Zhong
- Department of Rheumatology and Immunology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Jixin Zhong, ; Xiaoquan Rao, ; Lingli Dong,
| |
Collapse
|
8
|
Known Cellular and Receptor Interactions of Animal and Human Coronaviruses: A Review. Viruses 2022; 14:v14020351. [PMID: 35215937 PMCID: PMC8878323 DOI: 10.3390/v14020351] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/03/2022] [Accepted: 02/05/2022] [Indexed: 12/12/2022] Open
Abstract
This article aims to review all currently known interactions between animal and human coronaviruses and their cellular receptors. Over the past 20 years, three novel coronaviruses have emerged that have caused severe disease in humans, including SARS-CoV-2 (severe acute respiratory syndrome virus 2); therefore, a deeper understanding of coronavirus host-cell interactions is essential. Receptor-binding is the first stage in coronavirus entry prior to replication and can be altered by minor changes within the spike protein-the coronavirus surface glycoprotein responsible for the recognition of cell-surface receptors. The recognition of receptors by coronaviruses is also a major determinant in infection, tropism, and pathogenesis and acts as a key target for host-immune surveillance and other potential intervention strategies. We aim to highlight the need for a continued in-depth understanding of this subject area following on from the SARS-CoV-2 pandemic, with the possibility for more zoonotic transmission events. We also acknowledge the need for more targeted research towards glycan-coronavirus interactions as zoonotic spillover events from animals to humans, following an alteration in glycan-binding capability, have been well-documented for other viruses such as Influenza A.
Collapse
|
9
|
Gao X, Le Y, Geng C, Jiang Z, Zhao G, Zhang P. DPP4 Is a Potential Prognostic Marker of Thyroid Carcinoma and a Target for Immunotherapy. Int J Endocrinol 2022; 2022:5181386. [PMID: 36467461 PMCID: PMC9715318 DOI: 10.1155/2022/5181386] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/24/2022] [Accepted: 11/12/2022] [Indexed: 11/27/2022] Open
Abstract
DPP4 (dipeptidyl peptidase 4) is expressed in many cancers, but the relationship between DPP4 and thyroid carcinoma (THCA) is incompletely understood. We aim to explore the expression of DPP4 in THCA and the correlation between DPP4 expression with the prognosis of THCA and antitumor immunity. We systematically analyzed data from The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), and Gene Expression Omnibus (GEO) databases and explored DPP4 expression, its impact on prognosis, and its relationship with antitumor immunity in THCA. Next, we collected 18 pairs of fresh THCA and adjacent paracancerous tissues and performed RT-qPCR to validate the DPP4 mRNA level. Concurrently, immunohistochemistry (IHC) analysis was performed on 12 pairs of paraffin-embedded tissues of medullary thyroid carcinoma (MTC) and paracancerous tissues to validate the DPP4 protein level. Bioinformatics analysis showed that DPP4 mRNA expression in THCA was significantly higher than that in paracancerous tissues (p < 0.01). DPP4 was expressed at the highest levels in MTC than in other pathological types. The DPP4 expression level was different between groups with different clinical characteristics. The higher the DPP4 expressed in THCA, the lower the disease-free survival (DFS) was (HR = 1.8, p=0.048). DPP4 was significantly correlated with immune cell infiltration and immune response and was positively associated with 21 immune checkpoint genes (ICGs) in THCA (p < 0.05). The results of RT-qPCR showed that the relative mRNA expression of DPP4 was significantly upregulated in 18 THCA tissues compared to that in paracancerous tissues (p=0.011). IHC results showed that the DPP4 protein level was higher in 12 MTC tissues than in paracancerous tissues (p=0.011). In conclusion, DPP4 is a potential prognostic marker of THCA and may become an effective target for immunotherapy.
Collapse
Affiliation(s)
- Xiaoqian Gao
- Department of Ultrasound, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China
| | - Yali Le
- Health Management Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China
| | - Chenchen Geng
- Department of Ultrasound, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China
| | - Zhen Jiang
- Department of Otorhinolaryngology-Head and Neck Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China
| | - Guanghui Zhao
- Medical Laboratory Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China
| | - Ping Zhang
- Department of Ultrasound, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China
- Health Management Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao 266035, China
| |
Collapse
|
10
|
Li S, Tao L, Yu X, Zheng H, Wu J, Hu F. Royal Jelly Proteins and Their Derived Peptides: Preparation, Properties, and Biological Activities. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:14415-14427. [PMID: 34807598 DOI: 10.1021/acs.jafc.1c05942] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Royal jelly, also called bee milk, is a source of high-quality proteins. Royal jelly proteins serve as not only a rich source of essential amino acids and functional donors but also an excellent substrate for preparing bioactive peptides. Most naturally occurring bioactive peptides in royal jelly are antibacterial, while peptides derived from proteolytic reactions are shown to exert antihypertensive, antioxidative, and anti-aging activities. Further studies are warranted to characterize the functional properties of major royal jelly proteins and peptides, to explore the preparation of bioactive peptides and the potential novel activities, to improve their bioavailability, to enhance the production efficiency for commercial availability, and finally to open up new applications for royal jelly as a functional food and potential therapeutic agent.
Collapse
Affiliation(s)
- Shanshan Li
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Lingchen Tao
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Xinyu Yu
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Huoqing Zheng
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Jianping Wu
- Department of Agricultural, Food and Nutritional Science, University of Alberta, 4-10 Ag/For Building, Edmonton, Alberta T6G 2P5, Canada
| | - Fuliang Hu
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, People's Republic of China
| |
Collapse
|
11
|
Kang SM, Park JH. Pleiotropic Benefits of DPP-4 Inhibitors Beyond Glycemic Control. CLINICAL MEDICINE INSIGHTS-ENDOCRINOLOGY AND DIABETES 2021; 14:11795514211051698. [PMID: 34733107 PMCID: PMC8558587 DOI: 10.1177/11795514211051698] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 09/20/2021] [Indexed: 12/14/2022]
Abstract
Dipeptidyl peptidase (DPP)-4 inhibitors are oral anti-diabetic medications that block the activity of the ubiquitous enzyme DPP-4. Inhibition of this enzyme increases the level of circulating active glucagon-like peptide (GLP)-1 secreted from L-cells in the small intestine. GLP-1 increases the glucose level, dependent on insulin secretion from pancreatic β-cells; it also decreases the abnormally increased level of glucagon, eventually decreasing the blood glucose level in patients with type 2 diabetes. DPP-4 is involved in many physiological processes other than the degradation of GLP-1. Therefore, the inhibition of DPP-4 may have numerous effects beyond glucose control. In this article, we review the pleiotropic effects of DPP-4 inhibitors beyond glucose control, including their strong beneficial effects on the stress induced accelerated senescence of vascular cells, and the possible clinical implications of these effects.
Collapse
Affiliation(s)
- Seon Mee Kang
- Department of Internal Medicine, College of Medicine, Inje University, Busan, Republic of Korea.,Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea
| | - Jeong Hyun Park
- Department of Internal Medicine, College of Medicine, Inje University, Busan, Republic of Korea.,Paik Institute for Clinical Research, Inje University, Busan, Republic of Korea
| |
Collapse
|
12
|
Li QK, Chen J, Hu Y, Höti N, Lih TSM, Thomas SN, Chen L, Roy S, Meeker A, Shah P, Chen L, Bova GS, Zhang B, Zhang H. Proteomic characterization of primary and metastatic prostate cancer reveals reduced proteinase activity in aggressive tumors. Sci Rep 2021; 11:18936. [PMID: 34556748 PMCID: PMC8460832 DOI: 10.1038/s41598-021-98410-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/03/2021] [Indexed: 12/29/2022] Open
Abstract
Prostate cancer (PCa) is a heterogeneous group of tumors with variable clinical courses. In order to improve patient outcomes, it is critical to clinically separate aggressive PCa (AG) from non-aggressive PCa (NAG). Although recent genomic studies have identified a spectrum of molecular abnormalities associated with aggressive PCa, it is still challenging to separate AG from NAG. To better understand the functional consequences of PCa progression and the unique features of the AG subtype, we studied the proteomic signatures of primary AG, NAG and metastatic PCa. 39 PCa and 10 benign prostate controls in a discovery cohort and 57 PCa in a validation cohort were analyzed using a data-independent acquisition (DIA) SWATH-MS platform. Proteins with the highest variances (top 500 proteins) were annotated for the pathway enrichment analysis. Functional analysis of differentially expressed proteins in NAG and AG was performed. Data was further validated using a validation cohort; and was also compared with a TCGA mRNA expression dataset and confirmed by immunohistochemistry (IHC) using PCa tissue microarray (TMA). 4,415 proteins were identified in the tumor and benign control tissues, including 158 up-regulated and 116 down-regulated proteins in AG tumors. A functional analysis of tumor-associated proteins revealed reduced expressions of several proteinases, including dipeptidyl peptidase 4 (DPP4), carboxypeptidase E (CPE) and prostate specific antigen (KLK3) in AG and metastatic PCa. A targeted analysis further identified that the reduced expression of DPP4 was associated with the accumulation of DPP4 substrates and the reduced ratio of DPP4 cleaved peptide to intact substrate peptide. Findings were further validated using an independently-collected tumor cohort, correlated with a TCGA mRNA dataset, and confirmed by immunohistochemical stains of PCa tumor microarray (TMA). Our study is the first large-scale proteomics analysis of PCa tissue using a DIA SWATH-MS platform. It provides not only an interrogative proteomic signature of PCa subtypes, but also indicates the critical roles played by certain proteinases during tumor progression. The spectrum map and protein profile generated in the study can be used to investigate potential biological mechanisms involved in PCa and for the development of a clinical assay to distinguish aggressive from indolent PCa.
Collapse
Affiliation(s)
- Qing Kay Li
- Department of Pathology, The John Hopkins Medical Institutions, 600 N. Wolfe Street, Baltimore, MD, 21224, USA.
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
| | - Jing Chen
- Department of Pathology, The John Hopkins Medical Institutions, 600 N. Wolfe Street, Baltimore, MD, 21224, USA
| | - Yingwei Hu
- Department of Pathology, The John Hopkins Medical Institutions, 600 N. Wolfe Street, Baltimore, MD, 21224, USA
| | - Naseruddin Höti
- Department of Pathology, The John Hopkins Medical Institutions, 600 N. Wolfe Street, Baltimore, MD, 21224, USA
| | - Tung-Shing Mamie Lih
- Department of Pathology, The John Hopkins Medical Institutions, 600 N. Wolfe Street, Baltimore, MD, 21224, USA
| | - Stefani N Thomas
- Department of Pathology, The John Hopkins Medical Institutions, 600 N. Wolfe Street, Baltimore, MD, 21224, USA
| | - Li Chen
- Department of Pathology, The John Hopkins Medical Institutions, 600 N. Wolfe Street, Baltimore, MD, 21224, USA
| | - Sujayita Roy
- Department of Pathology, The John Hopkins Medical Institutions, 600 N. Wolfe Street, Baltimore, MD, 21224, USA
| | - Alan Meeker
- Department of Pathology, The John Hopkins Medical Institutions, 600 N. Wolfe Street, Baltimore, MD, 21224, USA
| | - Punit Shah
- Department of Pathology, The John Hopkins Medical Institutions, 600 N. Wolfe Street, Baltimore, MD, 21224, USA
| | - Lijun Chen
- Department of Pathology, The John Hopkins Medical Institutions, 600 N. Wolfe Street, Baltimore, MD, 21224, USA
| | - G Steven Bova
- Prostate Cancer Research Center, Faculty of Medicine and Health Technology, Tampere University, FI-33014, Tampere, Finland
| | - Bai Zhang
- Department of Pathology, The John Hopkins Medical Institutions, 600 N. Wolfe Street, Baltimore, MD, 21224, USA
| | - Hui Zhang
- Department of Pathology, The John Hopkins Medical Institutions, 600 N. Wolfe Street, Baltimore, MD, 21224, USA.
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
- Department of Urology, Sidney Kimmel Cancer Center, Johns Hopkins Medical Institutions, Baltimore, MD, USA.
- Johns Hopkins University, 400 N. Broadway, Smith Bldg Rm 4011, Baltimore, MD, 21287, USA.
| |
Collapse
|
13
|
De Zutter A, Van Damme J, Struyf S. The Role of Post-Translational Modifications of Chemokines by CD26 in Cancer. Cancers (Basel) 2021; 13:cancers13174247. [PMID: 34503058 PMCID: PMC8428238 DOI: 10.3390/cancers13174247] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/04/2021] [Accepted: 08/10/2021] [Indexed: 02/06/2023] Open
Abstract
Chemokines are a large family of small chemotactic cytokines that fulfill a central function in cancer. Both tumor-promoting and -impeding roles have been ascribed to chemokines, which they exert in a direct or indirect manner. An important post-translational modification that regulates chemokine activity is the NH2-terminal truncation by peptidases. CD26 is a dipeptidyl peptidase (DPPIV), which typically clips a NH2-terminal dipeptide from the chemokine. With a certain degree of selectivity in terms of chemokine substrate, CD26 only recognizes chemokines with a penultimate proline or alanine. Chemokines can be protected against CD26 recognition by specific amino acid residues within the chemokine structure, by oligomerization or by binding to cellular glycosaminoglycans (GAGs). Upon truncation, the binding affinity for receptors and GAGs is altered, which influences chemokine function. The consequences of CD26-mediated clipping vary, as unchanged, enhanced, and reduced activities are reported. In tumors, CD26 most likely has the most profound effect on CXCL12 and the interferon (IFN)-inducible CXCR3 ligands, which are converted into receptor antagonists upon truncation. Depending on the tumor type, expression of CD26 is upregulated or downregulated and often results in the preferential generation of the chemokine isoform most favorable for tumor progression. Considering the tight relationship between chemokine sequence and chemokine binding specificity, molecules with the appropriate characteristics can be chemically engineered to provide innovative therapeutic strategies in a cancer setting.
Collapse
|
14
|
Khursheed A, Jain V, Rasool A, Rather MA, Malik NA, Shalla AH. Molecular scaffolds from mother nature as possible lead compounds in drug design and discovery against coronaviruses: A landscape analysis of published literature and molecular docking studies. Microb Pathog 2021; 157:104933. [PMID: 33984466 PMCID: PMC8110334 DOI: 10.1016/j.micpath.2021.104933] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 04/13/2021] [Accepted: 04/28/2021] [Indexed: 12/23/2022]
Abstract
The recent outbreak of viral infection and its transmission has highlighted the importance of its slowdown for the safeguard of public health, globally. The identification of novel drugs and efficient therapies against these infectious viruses is need of the hour. The eruption of COVID-19 is caused by a novel acute respiratory syndrome virus SARS-CoV-2 which has taken the whole world by storm as it has transformed into a global pandemic. This lethal syndrome is a global health threat to general public which has already affected millions of people. Despite the development of some potential vaccines and repurposed drugs by some Pharma companies, this health emergency needs more attention due to the less efficacy of these vaccines coupled with the emergence of novel and resistant strains of SARS-CoV-2. Due to enormous structural diversity and biological applications, natural products are considered as a wonderful source of drugs for such diseases. Natural product based drugs constitute a substantial proportion of the pharmaceutical market particularly in the therapeutic areas of infectious diseases and oncology. The naturally occurring bioactive antiviral phytochemicals including alkaloids, flavonoids and peptides have been subjected to virtual screening against COVID-19. Since there is no specific medicine available for the treatment of Covid-19, designing new drugs using in silico methods plays an all important role to find that magic bullet which can target this lethal virus. The in silico method is not only quick but economical also when compared to the other conventional methods which are hit and trial methods. Based on this in silico approach, various natural products have been recently identified which might have a potential to inhibit COVID-19 outbreak. These natural products have been shown by these docking studies to interact with the spike protein of the novel coronavirus. This spike protein has been shown to bind to a transmembrane protein called Angiotensin converting enzyme 2 (ACE2), this protein acts as a receptor for the viral spike protein. This comprehensive review article anticipates providing a summary of the authentic and peer reviewed published literature about the potential of natural metabolites that can be developed into possible lead compounds against this new threat of Covid-19. Main focus of the article will be to highlight natural sources of potential anti-coronavirus molecules, mechanism of action, docking studies and the target proteins as well as their toxicity profiles. This review article intends to provide a starting point for the research endeavors that are needed for the design and development of drugs based on pure natural products, their synthetic or semi-synthetic derivatives and standardized plant extracts. This review article will be highly helpful for scientists who are working or intend to work on antiviral drugs from natural sources.
Collapse
Affiliation(s)
- Aadil Khursheed
- Department of Chemistry, Madhyanchal Professional University, Ratibad, Bhopal, 462044, Madhya Pradesh, India
| | - Vikrant Jain
- Department of Chemistry, Madhyanchal Professional University, Ratibad, Bhopal, 462044, Madhya Pradesh, India
| | - Ajaz Rasool
- Department of Zoology, University of Kashmir, Srinagar, 190006, India
| | - Manzoor A Rather
- Department of Chemistry, Islamic University of Science and Technology, Awanti Pora, 192122, Jammu and Kashmir, India.
| | - Nisar Ahmad Malik
- Department of Chemistry, Islamic University of Science and Technology, Awanti Pora, 192122, Jammu and Kashmir, India
| | - Aabid Hussain Shalla
- Department of Chemistry, Islamic University of Science and Technology, Awanti Pora, 192122, Jammu and Kashmir, India
| |
Collapse
|
15
|
Rey F. Structure-function relations of the SARS-CoV-2 spike protein and impact of mutations in the variants of concern. C R Biol 2021; 344:77-110. [PMID: 34213849 DOI: 10.5802/crbiol.53] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This review covers the main features of the severe acquired respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein, its interaction with the main entry receptor, the human angiotensin converting enzyme 2 (ACE2), and the subsequent membrane fusion step. The focus is on the structural organization of these proteins and mechanistic aspects of their interactions that lead to cytoplasmic release of the viral genome. The most potently neutralizing antibodies against SARS-CoV-2 were shown to interfere with the spike/ACE2 interaction. I thus also review the location and the potential impact of mutations in the spike protein observed in the variants of concern that emerged concomitantly with acquired immunity in the population after one year of virus circulation. Understanding how these interactions affect the spike/ACE2 interactions and the subsequent spike-protein-induced membrane fusion reaction is important to stay one step ahead of the virus evolution and develop efficient countermeasures.
Collapse
Affiliation(s)
- Félix Rey
- Unité de Virologie Structurale, CNRS UMR 3569, Institut Pasteur, 28 rue du Dr Roux, 75015 Paris, France
| |
Collapse
|
16
|
Kawakita E, Koya D, Kanasaki K. CD26/DPP-4: Type 2 Diabetes Drug Target with Potential Influence on Cancer Biology. Cancers (Basel) 2021; 13:cancers13092191. [PMID: 34063285 PMCID: PMC8124456 DOI: 10.3390/cancers13092191] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Dipeptidyl peptidase (DPP)-4 inhibitor is widely used for type 2 diabetes. Although DPP-4/CD26 has been recognized as both a suppressor and inducer in tumor biology due to its various functions, how DPP-4 inhibitor affects cancer progression in diabetic patients is still unknown. The aim of this review is to summarize one unfavorable aspect of DPP-4 inhibitor in cancer-bearing diabetic patients. Abstract DPP-4/CD26, a membrane-bound glycoprotein, is ubiquitously expressed and has diverse biological functions. Because of its enzymatic action, such as the degradation of incretin hormones, DPP-4/CD26 is recognized as the significant therapeutic target for type 2 diabetes (T2DM); DPP-4 inhibitors have been used as an anti-diabetic agent for a decade. The safety profile of DPP-4 inhibitors for a cardiovascular event in T2DM patients has been widely analyzed; however, a clear association between DPP-4 inhibitors and tumor biology is not yet established. Previous preclinical studies reported that DPP-4 suppression would impact tumor progression processes. With regard to this finding, we have shown that the DPP-4 inhibitor induces breast cancer metastasis and chemoresistance via an increase in its substrate C-X-C motif chemokine 12, and the consequent induction of epithelial-mesenchymal transition in the tumor. DPP-4/CD26 plays diverse pivotal roles beyond blood glucose control; thus, DPP-4 inhibitors can potentially impact cancer-bearing T2DM patients either favorably or unfavorably. In this review, we primarily focus on the possible undesirable effect of DPP-4 inhibition on tumor biology. Clinicians should note that the safety of DPP-4 inhibitors for diabetic patients with an existing cancer is an unresolved issue, and further mechanistic analysis is essential in this field.
Collapse
Affiliation(s)
- Emi Kawakita
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo 693-8501, Japan;
| | - Daisuke Koya
- Department of Diabetology & Endocrinology, Kanazawa Medical University, Uchinada 920-0293, Japan;
- Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Japan
| | - Keizo Kanasaki
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1 Enya-cho, Izumo 693-8501, Japan;
- Division of Anticipatory Molecular Food Science and Technology, Medical Research Institute, Kanazawa Medical University, Uchinada 920-0293, Japan
- Correspondence: ; Tel.: +81-853-20-2183
| |
Collapse
|
17
|
Knura M, Garczorz W, Borek A, Drzymała F, Rachwał K, George K, Francuz T. The Influence of Anti-Diabetic Drugs on Prostate Cancer. Cancers (Basel) 2021; 13:cancers13081827. [PMID: 33921222 PMCID: PMC8068793 DOI: 10.3390/cancers13081827] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/27/2021] [Accepted: 04/08/2021] [Indexed: 12/13/2022] Open
Abstract
The incidences of prostate cancer (PC) and diabetes are increasing, with a sustained trend. The occurrence of PC and type 2 diabetes mellitus (T2DM) is growing with aging. The correlation between PC occurrence and diabetes is noteworthy, as T2DM is correlated with a reduced risk of incidence of prostate cancer. Despite this reduction, diabetes mellitus increases the mortality in many cancer types, including prostate cancer. The treatment of T2DM is based on lifestyle changes and pharmacological management. Current available drugs, except insulin, are aimed at increasing insulin secretion (sulfonylureas, incretin drugs), improving insulin sensitivity (biguanides, thiazolidinediones), or increasing urinary glucose excretion (gliflozin). Comorbidities should be taken into consideration during the treatment of T2DM. This review describes currently known information about the mechanism and impact of commonly used antidiabetic drugs on the incidence and progression of PC. Outcomes of pre-clinical studies are briefly presented and their correlations with available clinical trials have also been observed. Available reports and meta-analyses demonstrate that most anti-diabetic drugs do not increase the risk during the treatment of patients with PC. However, some reports show a potential advantage of treatment of T2DM with specific drugs. Based on clinical reports, use of metformin should be considered as a therapeutic option. Moreover, anticancer properties of metformin were augmented while combined with GLP-1 analogs.
Collapse
|
18
|
Pan K, Ohnuma K, Morimoto C, Dang NH. CD26/Dipeptidyl Peptidase IV and Its Multiple Biological Functions. Cureus 2021; 13:e13495. [PMID: 33777580 PMCID: PMC7990348 DOI: 10.7759/cureus.13495] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
CD26/Dipeptidyl peptidase IV (DPPIV) is a cell surface glycoprotein with numerous roles including glucose metabolism, immunomodulation, and tumorigenesis. CD26/DPPIV is well recognized in diabetes, with DPPIV inhibitors being a class of oral hypoglycemic drugs called gliptins that are commonly used to treat type two diabetes mellitus. Recent work also indicated a potential role for CD26 in infectious diseases, including COVID-19, and immune-mediated disorders such as rheumatoid arthritis, inflammatory bowel disease, and graft-versus-host disease. In cancer, CD26/DPPIV expression has been characterized in numerous tumors such as hematologic malignancies, malignant pleural mesothelioma (MPM), renal cell carcinoma (RCC), hepatocellular carcinoma (HCC), gastrointestinal stromal tumor (GIST), and prostate, lung, colorectal, and ovarian (PLCO) cancer. Hence, CD26 has been frequently studied as a tumor biomarker and therapeutic target. CD26/DPPIV-targeted therapies have been evaluated in various cancers, including the use of anti-CD26 monoclonal antibodies as anticancer treatment in selected neoplasms. This review highlights our current understanding of the role of CD26 in cancer, diabetes, immune-mediated diseases, and infectious diseases. Enhanced understanding of CD26 biology and function may lead to novel therapeutic approaches in multiple human diseases.
Collapse
Affiliation(s)
- Kelsey Pan
- Internal Medicine, University of Florida, Gainesville, USA
| | - Kei Ohnuma
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Juntendo University, Tokyo, JPN
| | - Chikao Morimoto
- Department of Therapy Development and Innovation for Immune Disorders and Cancers, Juntendo University, Tokyo, JPN
| | - Nam H Dang
- Oncology, University of Florida, Gainesville, USA
| |
Collapse
|
19
|
Chaudhary MK, Karthick T, Joshi BD, Prajapati P, de Santana MSA, Ayala AP, Reeda VSJ, Tandon P. Molecular structure and quantum descriptors of cefradine by using vibrational spectroscopy (IR and Raman), NBO, AIM, chemical reactivity and molecular docking. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 246:118976. [PMID: 33017794 DOI: 10.1016/j.saa.2020.118976] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 08/21/2020] [Accepted: 09/18/2020] [Indexed: 06/11/2023]
Abstract
This study aims to investigate the structural and vibrational features of cefradine (the first-generation cephalosporin antibiotic) based on spectroscopic experiments and theoretical quantum chemical approach. The fundamental structural aspects of cefradine have been examined based on optimized geometry, spectroscopic behavior, intermolecular interaction, chemical reactivity, intramolecular hydrogen bonding, and molecular docking analysis. The most stable minimum energy conformer of the title molecule was identified by performing a one-dimensional potential energy surface scan along the rotational bonds at B3LYP/6-311++G (d,p) level of theory. The vibrational features of the molecule and information about the coupled modes were predicted. The chemical reactivity and stability of all the possible conformers of cefradine were estimated based on the HOMO-LUMO energy gap and NBO approach. The overall picture of accumulation of charges on individual atoms of the molecule was predicted by molecular electrostatic potential (MEP) surface map which in turn identifies the nucleophilic and electrophilic region or sites. The quantitative analysis of electrophilicity and nucleophilicity indices was done by Hirshfeld charge analysis and it was found that N8 atom is the most prominent site for nucleophilic attack while C14 atom is feasible for electrophilic attack. QTAIM study has also been performed to investigate the nature and strength of hydrogen bonding interactions. Besides, molecular docking studies were performed to examine the active binding residues of the target.
Collapse
Affiliation(s)
- Manoj Kumar Chaudhary
- Central Department of Physics, Tribhuvan University, Kirtipur, Kathmandu, Nepal; Department of Physics, University of Lucknow, Lucknow 226 007, India
| | - T Karthick
- Department of Physics, School of Electrical and Electronics Engineering, SASTRA Deemed University, Thanjavur 613 401, Tamil Nadu, India
| | - Bhawani Datt Joshi
- Department of Physics, Siddhanath Science Campus, Tribhuvan University, Mahendranagar 10406, Nepal.
| | - Preeti Prajapati
- Department of Physics, University of Lucknow, Lucknow 226 007, India
| | | | - Alejandro Pedro Ayala
- Depertmento de Fisica, Universidade Federal do Ceará, C.P. 6030,60.455-900, Fortaleza, CE, Brazil
| | - V S Jeba Reeda
- Department of Physics and Research Center, Women's Christian College, Nagercoil 629001, Tamil Nadu, India
| | - Poonam Tandon
- Department of Physics, University of Lucknow, Lucknow 226 007, India.
| |
Collapse
|
20
|
Manocha E, Bugatti A, Belleri M, Zani A, Marsico S, Caccuri F, Presta M, Caruso A. Avian Reovirus P17 Suppresses Angiogenesis by Promoting DPP4 Secretion. Cells 2021; 10:cells10020259. [PMID: 33525607 PMCID: PMC7911508 DOI: 10.3390/cells10020259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/18/2021] [Accepted: 01/22/2021] [Indexed: 12/17/2022] Open
Abstract
Avian reovirus p17 (ARV p17) is a non-structural protein known to activate autophagy, interfere with gene transcription and induce a significant tumor cell growth inhibition in vitro and in vivo. In this study, we show that ARV p17 is capable of exerting potent antiangiogenic properties. The viral protein significantly inhibited the physiological angiogenesis of human endothelial cells (ECs) by affecting migration, capillary-like structure and new vessel formation. ARV p17 was not only able to suppress the EC physiological angiogenesis but also rendered ECs insensitive to two different potent proangiogenic inducers, such as VEGF-A and FGF-2 in the three-dimensional (3D) Matrigel and spheroid assay. ARV p17 was found to exert its antiangiogenic activity by upregulating transcription and release of the well-known tumor suppressor molecule dipeptidyl peptidase 4 (DPP4). The ability of ARV p17 to impact on angiogenesis is completely new and highlights the “two compartments” activity of the viral protein that is expected to hamper the tumor parenchymal/stromal crosstalk. The complex antitumor activities of ARV p17 open the way to a new promising field of research aimed to develop new therapeutic approaches for treating tumor and cancer metastasis.
Collapse
Affiliation(s)
- Ekta Manocha
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (E.M.); (A.B.); (A.Z.); (F.C.)
| | - Antonella Bugatti
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (E.M.); (A.B.); (A.Z.); (F.C.)
| | - Mirella Belleri
- Section of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.B.); (M.P.)
| | - Alberto Zani
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (E.M.); (A.B.); (A.Z.); (F.C.)
| | - Stefania Marsico
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy;
| | - Francesca Caccuri
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (E.M.); (A.B.); (A.Z.); (F.C.)
| | - Marco Presta
- Section of Experimental Oncology and Immunology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (M.B.); (M.P.)
| | - Arnaldo Caruso
- Section of Microbiology, Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy; (E.M.); (A.B.); (A.Z.); (F.C.)
- Correspondence:
| |
Collapse
|
21
|
Systematic Analysis of the Aberrances and Functional Implications of Ferroptosis in Cancer. iScience 2020; 23:101302. [PMID: 32629423 PMCID: PMC7334617 DOI: 10.1016/j.isci.2020.101302] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/09/2020] [Accepted: 06/17/2020] [Indexed: 12/12/2022] Open
Abstract
Ferroptosis is a type of cell death related to cancer; however, the characteristics of ferroptosis in cancers are still uncertain. Based on the data in The Cancer Genome Atlas, we found that most ferroptosis regulator genes (FRGs) were differentially expressed in tumors, somatic copy number alterations (SCNA) and DNA methylation contributed to their aberrant expression. We established the ferroptosis potential index (FPI) to reveal the functional roles of ferroptosis and noticed that the FPI was higher in tumors than in normal tissues in most cancers and was associated with subtypes and clinical features. The FPI was negatively correlated with several metabolic pathways but positively associated with several important metastasis-related pathways and immune-related pathways. High FPI predicted poor prognosis in several tumors, whereas FPI and FRGs impacted drug sensitivity. Our study presents a systematic analysis of ferroptosis and its regulatory genes and highlights the potential of ferroptosis-based cancer therapy. The ferroptosis regulator genes were aberrantly expressed in tumor The ferroptosis potential index (FPI) was established to model ferroptosis level The FPI was correlated with metabolic, metastatic, and immune pathways High FPI predicted poor prognosis in many cancer types
Collapse
|
22
|
Shao S, Xu Q, Yu X, Pan R, Chen Y. Dipeptidyl peptidase 4 inhibitors and their potential immune modulatory functions. Pharmacol Ther 2020; 209:107503. [PMID: 32061923 PMCID: PMC7102585 DOI: 10.1016/j.pharmthera.2020.107503] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 01/30/2020] [Indexed: 12/25/2022]
Abstract
Dipeptidyl peptidase 4 (DPP4) inhibitors (DPP4is) are oral anti-diabetic drugs (OADs) for the treatment of type 2 diabetes mellitus (T2DM) through inhibiting the degradation of incretin peptides. Numerous investigations have been focused on the effects of DPP4is on glucose homeostasis. However, there are limited evidences demonstrating their Potential modulatory functions in the immune system. DPP4, originally known as the lymphocyte cell surface protein CD26, is widely expressed in many types of immune cells including CD4(+) and CD8(+) T cells, B cells, NK cells, dendritic cells, and macrophages; and regulate the functions of these cells. In addition, DPP4 is capable of modulating plenty of cytokines, chemokines and peptide hormones. Accordingly, DPP4/CD26 is speculated to be involved in various immune/inflammatory diseases and DPP4is may become a new drug class applied in these diseases. This review focuses on the regulatory effects of DPP4is on immune functions and their possible underlying mechanisms. Further clinical studies will be necessitated to fully evaluate the administration of DPP4is in diabetic patients with or without immune diseases.
Collapse
Affiliation(s)
- Shiying Shao
- Division of Endocrinology, Department of Internal Medicine, Tongji hospital, Tongji medical college, Huazhong University of Science & Technology, Wuhan 430030, PR China
| | - QinQin Xu
- Division of Endocrinology, Department of Internal Medicine, Tongji hospital, Tongji medical college, Huazhong University of Science & Technology, Wuhan 430030, PR China
| | - Xuefeng Yu
- Division of Endocrinology, Department of Internal Medicine, Tongji hospital, Tongji medical college, Huazhong University of Science & Technology, Wuhan 430030, PR China
| | - Ruping Pan
- Department of Nuclear Medicine, Tongji hospital, Tongji medical college, Huazhong University of Science & Technology, Wuhan 430030, PR China
| | - Yong Chen
- Division of Endocrinology, Department of Internal Medicine, Tongji hospital, Tongji medical college, Huazhong University of Science & Technology, Wuhan 430030, PR China.
| |
Collapse
|
23
|
Li L, van Breugel PC, Loayza-Puch F, Ugalde AP, Korkmaz G, Messika-Gold N, Han R, Lopes R, Barbera EP, Teunissen H, de Wit E, Soares RJ, Nielsen BS, Holmstrøm K, Martínez-Herrera DJ, Huarte M, Louloupi A, Drost J, Elkon R, Agami R. LncRNA-OIS1 regulates DPP4 activation to modulate senescence induced by RAS. Nucleic Acids Res 2019; 46:4213-4227. [PMID: 29481642 PMCID: PMC5934637 DOI: 10.1093/nar/gky087] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/29/2018] [Indexed: 12/15/2022] Open
Abstract
Oncogene-induced senescence (OIS), provoked in response to oncogenic activation, is considered an important tumor suppressor mechanism. Long non-coding RNAs (lncRNAs) are transcripts longer than 200 nt without a protein-coding capacity. Functional studies showed that deregulated lncRNA expression promote tumorigenesis and metastasis and that lncRNAs may exhibit tumor-suppressive and oncogenic function. Here, we first identified lncRNAs that were differentially expressed between senescent and non-senescent human fibroblast cells. Using RNA interference, we performed a loss-function screen targeting the differentially expressed lncRNAs, and identified lncRNA-OIS1 (lncRNA#32, AC008063.3 or ENSG00000233397) as a lncRNA required for OIS. Knockdown of lncRNA-OIS1 triggered bypass of senescence, higher proliferation rate, lower abundance of the cell-cycle inhibitor CDKN1A and high expression of cell-cycle-associated genes. Subcellular inspection of lncRNA-OIS1 indicated nuclear and cytosolic localization in both normal culture conditions as well as following oncogene induction. Interestingly, silencing lncRNA-OIS1 diminished the senescent-associated induction of a nearby gene (Dipeptidyl Peptidase 4, DPP4) with established role in tumor suppression. Intriguingly, similar to lncRNA-OIS1, silencing DPP4 caused senescence bypass, and ectopic expression of DPP4 in lncRNA-OIS1 knockdown cells restored the senescent phenotype. Thus, our data indicate that lncRNA-OIS1 links oncogenic induction and senescence with the activation of the tumor suppressor DPP4.
Collapse
Affiliation(s)
- Li Li
- Division of Oncogenomics, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Pieter C van Breugel
- Division of Oncogenomics, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Fabricio Loayza-Puch
- Division of Oncogenomics, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Alejandro Pineiro Ugalde
- Division of Oncogenomics, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Gozde Korkmaz
- Division of Oncogenomics, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Naama Messika-Gold
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, 69978, Tel Aviv University, Tel Aviv, Israel
| | - Ruiqi Han
- Division of Oncogenomics, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Rui Lopes
- Division of Oncogenomics, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Eric P Barbera
- Division of Molecular Genetics, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Hans Teunissen
- Division of Gene Regulation, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Elzo de Wit
- Division of Gene Regulation, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | | | | | - Kim Holmstrøm
- Bioneer A/S, Kogle Allé 2, DK-2970 Hørsholm, Denmark
| | | | - Maite Huarte
- Institute of Health Research of Navarra (IdiSNA), 31008 Pamplona, Spain
| | - Annita Louloupi
- Division of Oncogenomics, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Jarno Drost
- Division of Oncogenomics, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Ran Elkon
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, 69978, Tel Aviv University, Tel Aviv, Israel
| | - Reuven Agami
- Division of Oncogenomics, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands.,Erasmus MC, Rotterdam University, 3000 CA Rotterdam, The Netherlands.,Oncode institute, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| |
Collapse
|
24
|
Nieto-Fontarigo JJ, González-Barcala FJ, San José E, Arias P, Nogueira M, Salgado FJ. CD26 and Asthma: a Comprehensive Review. Clin Rev Allergy Immunol 2019; 56:139-160. [PMID: 27561663 PMCID: PMC7090975 DOI: 10.1007/s12016-016-8578-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Asthma is a heterogeneous and chronic inflammatory family of disorders of the airways with increasing prevalence that results in recurrent and reversible bronchial obstruction and expiratory airflow limitation. These diseases arise from the interaction between environmental and genetic factors, which collaborate to cause increased susceptibility and severity. Many asthma susceptibility genes are linked to the immune system or encode enzymes like metalloproteases (e.g., ADAM-33) or serine proteases. The S9 family of serine proteases (prolyl oligopeptidases) is capable to process peptide bonds adjacent to proline, a kind of cleavage-resistant peptide bonds present in many growth factors, chemokines or cytokines that are important for asthma. Curiously, two serine proteases within the S9 family encoded by genes located on chromosome 2 appear to have a role in asthma: CD26/dipeptidyl peptidase 4 (DPP4) and DPP10. The aim of this review is to summarize the current knowledge about CD26 and to provide a structured overview of the numerous functions and implications that this versatile enzyme could have in this disease, especially after the detection of some secondary effects (e.g., viral nasopharyngitis) in type II diabetes mellitus patients (a subset with a certain risk of developing obesity-related asthma) upon CD26 inhibitory therapy.
Collapse
Affiliation(s)
- Juan J Nieto-Fontarigo
- Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Francisco J González-Barcala
- Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
- Respiratory Department, Clinic University Hospital (CHUS), Santiago de Compostela, Spain
| | - Esther San José
- Clinical Analysis Service, Clinic University Hospital (CHUS), Santiago de Compostela, Spain
| | - Pilar Arias
- Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Montserrat Nogueira
- Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Francisco J Salgado
- Department of Biochemistry and Molecular Biology, Faculty of Biology-Biological Research Centre (CIBUS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain.
| |
Collapse
|
25
|
Shao S, Wang C, Tian J, Zhang H, Wang S, Du Y. Diagnostic and prognostic significance of serum CD26 level in Asian women with high-grade serous ovarian carcinoma. Future Oncol 2019; 15:1863-1871. [PMID: 31140312 DOI: 10.2217/fon-2018-0725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The aim of this study was to reveal the diagnostic and prognostic significance of serum CD26 level in high-grade serous ovarian carcinoma women in China. Methods: There were 229 high-grade serous ovarian carcinoma women and 365 controls. Baseline serum CD26 level was measured using ELISA. A 36-month post-operation follow-up was performed. Results: Baseline serum CD26 level ≤601.5 pg/ml was associated with the increased risk of ovarian carcinoma (OR: 1.67; 95% CI: 1.20-2.32). Baseline serum level of CD26 ≤589.7 pg/ml was related to the elevated risk of cancer death (HR: 1.33; 95% CI: 1.04-1.69). Conclusion: Baseline serum CD26 level might be an independent diagnostic and prognostic marker for high-grade serous ovarian carcinoma.
Collapse
Affiliation(s)
- Shiqing Shao
- Department of Obstetrics & Gynecology, Huaihe Hospital, Henan University, Kaifeng, 475000, Henan Province, PR China
| | - Chen Wang
- Department of Obstetrics & Gynecology, Huaihe Hospital, Henan University, Kaifeng, 475000, Henan Province, PR China
| | - Jun Tian
- Department of Obstetrics & Gynecology, Huaihe Hospital, Henan University, Kaifeng, 475000, Henan Province, PR China
| | - Hongxia Zhang
- Department of Obstetrics & Gynecology, Huaihe Hospital, Henan University, Kaifeng, 475000, Henan Province, PR China
| | - Shelian Wang
- Department of Obstetrics & Gynecology, Huaihe Hospital, Henan University, Kaifeng, 475000, Henan Province, PR China
| | - Yaowu Du
- Laboratory for Nanomedicine, School of Basic Medical Science, Henan University, Kaifeng, 475004, Henan Province, PR China
| |
Collapse
|
26
|
Enz N, Vliegen G, De Meester I, Jungraithmayr W. CD26/DPP4 - a potential biomarker and target for cancer therapy. Pharmacol Ther 2019; 198:135-159. [PMID: 30822465 DOI: 10.1016/j.pharmthera.2019.02.015] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CD26/dipeptidyl peptidase (DPP)4 is a membrane-bound protein found in many cell types of the body, and a soluble form is present in body fluids. There is longstanding evidence that various primary tumors and also metastases express CD26/DPP4 to a variable extent. By cleaving dipeptides from peptides with a proline or alanine in the penultimate position at the N-terminus, it regulates the activity of incretin hormones, chemokines and many other peptides. Due to these effects and interactions with other molecules, a tumor promoting or suppressing role can be attributed to CD26/DPP4. In this review, we discuss the existing evidence on the expression of soluble or membrane-bound CD26/DPP4 in malignant diseases, along with the most recent findings on CD26/DPP4 as a therapeutic target in specific malignancies. The expression and possible involvement of the related DPP8 and DPP9 in cancer are also reviewed. A higher expression of CD26/DPP4 is found in a wide variety of tumor entities, however more research on CD26/DPP4 in the tumor microenvironment is needed to fully explore its use as a tumor biomarker. Circulating soluble CD26/DPP4 has also been studied as a cancer biomarker, however, the observed decrease in most cancer patients does not seem to be cancer specific. Encouraging results from experimental work and a recently reported first phase clinical trial targeting CD26/DPP4 in mesothelioma, renal and urological tumors pave the way for follow-up clinical studies, also in other tumor entities, possibly leading to the development of more effective complementary therapies against cancer.
Collapse
Affiliation(s)
- Njanja Enz
- Department of Thoracic Surgery, University Hospital Rostock, Schillingallee 35, 18057 Rostock, Germany
| | - Gwendolyn Vliegen
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | - Ingrid De Meester
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium.
| | - Wolfgang Jungraithmayr
- Department of Thoracic Surgery, University Hospital Rostock, Schillingallee 35, 18057 Rostock, Germany.
| |
Collapse
|
27
|
Park DS, Kim K, Jang M, Choi SC. Role of dipeptidyl peptidase-4 as a potentiator of activin/nodal signaling pathway. BMB Rep 2018. [PMID: 30463640 PMCID: PMC6330939 DOI: 10.5483/bmbrep.2018.51.12.210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
DPP4 (dipeptidyl peptidase-4), a highly conserved transmembrane glycoprotein with an exo-peptidase activity, has been shown to contribute to glucose metabolism, immune regulation, signal transduction, and cell differentiation. Here, we show that DPP4 is involved in control of activin/nodal signaling in Xenopus early development. In support of this, gain of function of DPP4 augmented Smad2 phosphorylation as well as expression of target genes induced by activin or nodal signal. In addition, Dpp4 and Xnr1 showed synergistic effect on induction of ectopic dorsal body axis, when co-injected at suboptimal doses in early embryos. Conversely, saxagliptin, a DPP4 inhibitor repressed activin induction of Smad2 phosphorylation. Notably, overexpression of Dpp4 disrupted specification of dorsal body axis of embryo, leading to malformed phenotypes such as spina bifida and a shortened and dorsally bent axis. Together, these results suggest that DPP4 functions as a potentiator of activin/nodal signaling pathway.
Collapse
Affiliation(s)
- Dong-Seok Park
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Kyuhee Kim
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Minjoo Jang
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Sun-Cheol Choi
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea
| |
Collapse
|
28
|
Russo JW, Gao C, Bhasin SS, Voznesensky OS, Calagua C, Arai S, Nelson PS, Montgomery B, Mostaghel EA, Corey E, Taplin ME, Ye H, Bhasin M, Balk SP. Downregulation of Dipeptidyl Peptidase 4 Accelerates Progression to Castration-Resistant Prostate Cancer. Cancer Res 2018; 78:6354-6362. [PMID: 30242112 DOI: 10.1158/0008-5472.can-18-0687] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 08/14/2018] [Accepted: 09/18/2018] [Indexed: 12/26/2022]
Abstract
The standard treatment for metastatic prostate cancer, androgen deprivation therapy (ADT), is designed to suppress androgen receptor (AR) activity. However, men invariably progress to castration-resistant prostate cancer (CRPC), and AR reactivation contributes to progression in most cases. To identify mechanisms that may drive CRPC, we examined a VCaP prostate cancer xenograft model as tumors progressed from initial androgen sensitivity prior to castration to castration resistance and then on to relapse after combined therapy with further AR-targeted drugs (abiraterone plus enzalutamide). AR activity persisted in castration-resistant and abiraterone/enzalutamide-resistant xenografts and was associated with increased expression of the AR gene and the AR-V7 splice variant. We then assessed expression of individual AR-regulated genes to identify those that persisted, thereby contributing to tumor growth, versus those that decreased and may therefore exhibit tumor suppressor activities. The most significantly decreased AR target gene was dipeptidyl peptidase 4 (DPP4), which encodes a membrane-anchored protein that cleaves dipeptides from multiple growth factors, resulting in their increased degradation. DPP4 mRNA and protein were also decreased in clinical CRPC cases, and inhibition of DPP4 with sitagliptin enhanced the growth of prostate cancer xenografts following castration. Significantly, DPP4 inhibitors are frequently used to treat type 2 diabetes as they increase insulin secretion. Together, these results implicate DPP4 as an AR-regulated tumor suppressor gene whose loss enhances growth factor activity and suggest that treatment with DPP4 inhibitors may accelerate emergence of resistance to ADT.Significance: These findings identify DPP4 as an AR-stimulated tumor suppressor gene that is downregulated during progression to castration-resistant prostate cancer, warning that treatment with DPP4 inhibitors, commonly used to treat type 2 diabetes, may accelerate prostate cancer progression following androgen deprivation therapy. Cancer Res; 78(22); 6354-62. ©2018 AACR.
Collapse
Affiliation(s)
- Joshua W Russo
- Department of Medicine and Cancer Center, Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| | - Ce Gao
- Department of Medicine, Bioinformatic and Systems Biology Unit, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Swati S Bhasin
- Department of Medicine, Bioinformatic and Systems Biology Unit, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Olga S Voznesensky
- Department of Medicine and Cancer Center, Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Carla Calagua
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Seiji Arai
- Department of Medicine and Cancer Center, Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
- Department of Urology, Gunma University Hospital, Maebashi, Gunma, Japan
| | - Peter S Nelson
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Bruce Montgomery
- University of Washington School of Medicine, Seattle, Washington
| | | | - Eva Corey
- University of Washington School of Medicine, Seattle, Washington
| | - Mary-Ellen Taplin
- Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Huihui Ye
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Manoj Bhasin
- Department of Medicine, Bioinformatic and Systems Biology Unit, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Steven P Balk
- Department of Medicine and Cancer Center, Hematology-Oncology Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
29
|
Yang X, Zhang X, Wu R, Huang Q, Jiang Y, Qin J, Yao F, Jin G, Zhang Y. DPPIV promotes endometrial carcinoma cell proliferation, invasion and tumorigenesis. Oncotarget 2018; 8:8679-8692. [PMID: 28060721 PMCID: PMC5352432 DOI: 10.18632/oncotarget.14412] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 12/05/2016] [Indexed: 01/05/2023] Open
Abstract
Dipeptidyl peptidase IV (DPPIV), also known as CD26, is a 110-kDa cell surface glycoprotein expressed in various tissues. DPPIV reportedly plays a direct role in the progression of several human malignancies. DPPIV specific inhibitors are employed as antidiabetics and could potentially be repurposed to enhance anti-tumor immunotherapies. In the present study, we investigated the correlation between DPPIV expression and tumor progression in endometrial carcinoma (EC). DPPIV overexpression altered cell morphology and stimulated cell proliferation, invasion and tumorigenesis in vitro and in vivo. These effects were abrogated by DPPIV knockdown or pharmacological inhibition using sitagliptin. DPPIV overexpression increased hypoxia-inducible factor 1a (HIF-1a) and vascular endothelial growth factor A (VEGFA) expression to promote HIF-1a-VEGFA signaling. Our results indicated that DPPIV accelerated endometrial carcinoma progression and that sitagliptin may be an effective anti-EC therapeutic.
Collapse
Affiliation(s)
- Xiaoqing Yang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, NanTong, Jiangsu 226006, People's Republic of China
| | - Xinhua Zhang
- Department of Anatomy, Nantong University, Nantong, JiangSu 226000, People's Republic of China
| | - Rongrong Wu
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, NanTong, Jiangsu 226006, People's Republic of China
| | - Qicheng Huang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, NanTong, Jiangsu 226006, People's Republic of China
| | - Yao Jiang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, NanTong, Jiangsu 226006, People's Republic of China
| | - Jianbing Qin
- Department of Anatomy, Nantong University, Nantong, JiangSu 226000, People's Republic of China
| | - Feng Yao
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, NanTong, Jiangsu 226006, People's Republic of China
| | - Guohua Jin
- Department of Anatomy, Nantong University, Nantong, JiangSu 226000, People's Republic of China
| | - Yuquan Zhang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, NanTong, Jiangsu 226006, People's Republic of China
| |
Collapse
|
30
|
Torrealba MP, Manfrere KC, Miyashiro DR, Lima JF, de M Oliveira L, Pereira NZ, Cury-Martins J, Pereira J, Duarte AJS, Sato MN, Sanches JA. Chronic activation profile of circulating CD8+ T cells in Sézary syndrome. Oncotarget 2017; 9:3497-3506. [PMID: 29423061 PMCID: PMC5790478 DOI: 10.18632/oncotarget.23334] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 12/01/2017] [Indexed: 12/13/2022] Open
Abstract
Sézary syndrome (SS) is a leukemic variant of cutaneous T cell lymphoma (CTCL), and the neoplastic CD4+ T cells of SS patients undergo intense clonal proliferation. Although Sézary cells have been studied extensively, studies on adaptive immunity regarding CD8+T cells are scarce. This study aimed to investigate activation marker expression in CD8+ T cells according to the differentiation stages and IL-7/IL7Rα axis responses of patients with SS. Moreover, this study aimed to verify the soluble forms of CD38, sCD127 and IL-7 in serum. Although the SS patients of our cohort had reduced numbers of CD8+ T cells, they exhibited higher percentages of CD8+CD38+ T cells, mainly effector/memory CD8+ T cells, than the control group. In contrast, down-regulated expression of the activation markers CD127/IL-7R and CD26 was found in the CD8+ T cells of SS patients. High serum levels of sCD38 and sCD127 and scarce serum levels of IL-7 were detected, emphasizing the immune activation status of SS patients. Moreover, CD8+ T cells from SS patients exhibited IL-7 unresponsiveness to STAT5 phosphorylation and Bcl-2 expression, and IL-7 priming partially restored IFNγ production. Our findings showed a chronic activation profile of CD8+ T cells, as an attenuated cytotoxic profile and impaired IL-7 responsiveness was observed, suggesting chronic activation status of CD8+ T cells in SS patients.
Collapse
Affiliation(s)
- Marina Passos Torrealba
- Medical Investigation Laboratory (LIM-56), Tropical Medicine Institute of São Paulo, Department of Dermatology, University of São Paulo Medical School, São Paulo, Brazil
| | - Kelly Cristina Manfrere
- Medical Investigation Laboratory (LIM-56), Tropical Medicine Institute of São Paulo, Department of Dermatology, University of São Paulo Medical School, São Paulo, Brazil
| | - Denis R Miyashiro
- Cutaneous Lymphoma Clinic, Hospital das Clinicas, Department of Dermatology, University of São Paulo Medical School, São Paulo, Brazil
| | - Josenilson F Lima
- Medical Investigation Laboratory (LIM-56), Tropical Medicine Institute of São Paulo, Department of Dermatology, University of São Paulo Medical School, São Paulo, Brazil
| | - Luana de M Oliveira
- Medical Investigation Laboratory (LIM-56), Tropical Medicine Institute of São Paulo, Department of Dermatology, University of São Paulo Medical School, São Paulo, Brazil
| | - Nátalli Z Pereira
- Medical Investigation Laboratory (LIM-56), Tropical Medicine Institute of São Paulo, Department of Dermatology, University of São Paulo Medical School, São Paulo, Brazil
| | - Jade Cury-Martins
- Cutaneous Lymphoma Clinic, Hospital das Clinicas, Department of Dermatology, University of São Paulo Medical School, São Paulo, Brazil
| | - Juliana Pereira
- Hematology Department, Hospital das Clinicas, University of São Paulo Medical School, São Paulo, Brazil
| | - Alberto J S Duarte
- Medical Investigation Laboratory (LIM-56), Tropical Medicine Institute of São Paulo, Department of Dermatology, University of São Paulo Medical School, São Paulo, Brazil
| | - Maria N Sato
- Medical Investigation Laboratory (LIM-56), Tropical Medicine Institute of São Paulo, Department of Dermatology, University of São Paulo Medical School, São Paulo, Brazil
| | - José A Sanches
- Cutaneous Lymphoma Clinic, Hospital das Clinicas, Department of Dermatology, University of São Paulo Medical School, São Paulo, Brazil
| |
Collapse
|
31
|
Wang H, Rojo de la Vega M, Zhang DD, Yu S, Zheng H. Response to comment on "NRF2 activation by antioxidant antidiabetic agents accelerates tumor metastasis". Sci Transl Med 2017; 8:349lr1. [PMID: 27464746 DOI: 10.1126/scitranslmed.aag1805] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/21/2016] [Indexed: 11/02/2022]
Abstract
Multiple factors may affect the outcome of diabetic patients with cancer treated with antioxidant antidiabetic agents.
Collapse
Affiliation(s)
- Hui Wang
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Monserrat Rojo de la Vega
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA.
| | - Shicang Yu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.
| | - Hongting Zheng
- Department of Endocrinology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China.
| |
Collapse
|
32
|
Sitagliptin may reduce prostate cancer risk in male patients with type 2 diabetes. Oncotarget 2017; 8:19057-19064. [PMID: 27661113 PMCID: PMC5386669 DOI: 10.18632/oncotarget.12137] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/06/2016] [Indexed: 12/19/2022] Open
Abstract
This retrospective cohort study evaluated the risk of prostate cancer associated with sitagliptin use in Taiwanese male patients with type 2 diabetes mellitus by using the reimbursement databases of the National Health Insurance. Male patients with newly diagnosed type 2 diabetes mellitus at an age ≥25 years between 1999 and 2010 were recruited. A total of 37,924 ever users of sitagliptin and 426,276 never users were followed until December 31, 2011. The treatment effect of sitagliptin (for ever versus never users, and for tertiles of cumulative duration of therapy) was estimated by Cox regression incorporated with the inverse probability of treatment weighting using propensity score. Analyses were also conducted in a 1:1 matched pair cohort based on 8 digits of propensity score. Results showed that during follow-up, 84 ever users and 2,549 never users were diagnosed of prostate cancer, representing an incidence of 140.74 and 240.17 per 100,000 person-years, respectively. The hazard ratio (95% confidence intervals) for ever users versus never users was 0.613 (0.493-0.763). The respective hazard ratio for the first, second, and third tertile of cumulative duration of sitagliptin use <5.9, 5.9-12.7 and >12.7 months was 0.853 (0.601-1.210), 0.840 (0.598-1.179) and 0.304 (0.191-0.483), respectively; and was 0.856 (0.603-1.214), 0.695 (0.475-1.016) and 0.410 (0.277-0.608) for cumulative dose <15,000, 15,000-33,600 and >33,600 mg, respectively. Findings were supported by analyses in the matched cohort. In conclusion, sitagliptin significantly reduces the risk of prostate cancer, especially when the cumulative duration is >12.7 months or the cumulative dose >33,600 mg.
Collapse
|
33
|
Dipeptidyl peptidase-4 inhibitors and cancer risk in patients with type 2 diabetes: a meta-analysis of randomized clinical trials. Sci Rep 2017; 7:8273. [PMID: 28811622 PMCID: PMC5557948 DOI: 10.1038/s41598-017-07921-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 07/03/2017] [Indexed: 01/21/2023] Open
Abstract
Some recent studies have suggested that the use of dipeptidyl peptidase-4 inhibitors (DPP4i) is associated with cancer development. However, some other studies suggest no such association. The aim of the present study was to evaluate the effect of DPP4i on the risk of developing cancers. The electronic databases PubMed, Medline, EMBASE, Web of Science and Cochrane Library and the clinical trial registry were searched for published and unpublished randomized clinical trials on humans. Eligible studies were RCTs conducted in patients with type 2 diabetes mellitus, comparing DPP4i with a placebo or other active drugs. A total of 72 trials with 35,768 and 33,319 patients enrolled for DPP4i and the comparison drugs, respectively. Overall, no significant associations were detected between the use of DPP4i and cancer development, in comparison with the use of other active drugs or placebo. The results were consistent across pre-defined subgroups stratified by type of DPP4i, type of cancer, drug for comparison, trial duration, or baseline characteristics. The results of this meta-analysis suggest that patients with type 2 diabetes treated with DPP4i do not have a higher risk of developing cancers than patients treated with a placebo or other drugs.
Collapse
|
34
|
Tuccori M, Convertino I, Galiulo MT, Marino A, Capogrosso-Sansone A, Blandizzi C. Diabetes drugs and the incidence of solid cancers: a survey of the current evidence. Expert Opin Drug Saf 2017; 16:1133-1148. [PMID: 28748718 DOI: 10.1080/14740338.2017.1361401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The evaluation of the relationship between the use of antidiabetic drug and the occurrence of cancer is extremely challenging, both from the clinical and pharmacoepidemiological standpoint. This narrative review described the current evidence supporting a relationship between the use of antidiabetic drugs and the incidence of solid cancers. Areas covered: Data from pharmacoepidemiological studies on cancer incidence were presented for the main antidiabetic drugs and drug classes, including human insulin and insulin analogues, metformin, sulfonylureas, glinides, alpha-glucosidase inhibitors, thiazolidinediones, incretin mimetics, and sodium glucose co-transporter 2 inhibitors. The relationship between the use of antidiabetics and the incidence of solid cancer was described in strata by any cancer and by organ-specific cancer and by drug and by drug classes. Information supporting biological evidence and putative mechanisms were also provided. Expert opinion: The history of exploration of the relationship between antidiabetic drugs and the risk of solid cancers has showed several issues. Unrecognized biases and misinterpretations of study results have had important consequences that delayed the identification of actual risk and benefits of the use of antidiabetic drugs associated with cancer occurrence or progression. The lesson learned from the past should address the future research in this area, since in the majority of cases findings are controversial and confirmatory studies are warranted.
Collapse
Affiliation(s)
- Marco Tuccori
- a Unit of Adverse Drug Reaction Monitoring , University Hospital of Pisa , Pisa , Italy
| | - Irma Convertino
- b Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Maria Teresa Galiulo
- b Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Alessandra Marino
- b Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | | | - Corrado Blandizzi
- a Unit of Adverse Drug Reaction Monitoring , University Hospital of Pisa , Pisa , Italy.,b Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| |
Collapse
|
35
|
Flores IL, Santos-Silva AR, Coletta RD, Leme AFP, Lopes MA. Low expression of angiotensinogen and dipeptidyl peptidase 1 in saliva of patients with proliferative verrucous leukoplakia. World J Clin Cases 2016; 4:356-363. [PMID: 27900324 PMCID: PMC5112355 DOI: 10.12998/wjcc.v4.i11.356] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/14/2016] [Accepted: 09/18/2016] [Indexed: 02/05/2023] Open
Abstract
AIM To elucidate the profile of the salivary proteome.
METHODS Unstimulated whole mouth saliva was collected from 30 volunteers [15 proliferative verrucous leukoplakia (PVL) patients and 15 controls] and proteins were submitted for mass spectrometry-based proteomics using the discovery approach, followed by analyses of variance and logistic regression tests.
RESULTS A total of two hundred and eighty-three proteins were confidently identified in saliva. By combining two low abundance proteins from the PVL group, angiotensinogen (AGT) and dipeptidyl peptidase 1 (DPP1), a model for group differentiation was built with a concordance index of 94.2%, identifying both proteins as potential etiologic biomarkers for PVL.
CONCLUSION This study suggests that both AGT and DPP1 may be involved in developmental mechanisms of PVL.
Collapse
|
36
|
Nakano T, Kuribayashi K, Mikami K. Possible new therapeutic agents for malignant pleural mesothelioma: anti-CD26 monoclonal antibody and naftopidil. Expert Rev Anticancer Ther 2016; 16:1097-1099. [PMID: 27718761 DOI: 10.1080/14737140.2016.1241150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Takashi Nakano
- a Division of Respiratory Diseases, Department of Internal Medicine , Hyogo College of Medicine , Hyogo , Japan
| | - Kozo Kuribayashi
- a Division of Respiratory Diseases, Department of Internal Medicine , Hyogo College of Medicine , Hyogo , Japan
| | - Koji Mikami
- a Division of Respiratory Diseases, Department of Internal Medicine , Hyogo College of Medicine , Hyogo , Japan
| |
Collapse
|
37
|
Abstract
The coronavirus spike protein is a multifunctional molecular machine that mediates coronavirus entry into host cells. It first binds to a receptor on the host cell surface through its S1 subunit and then fuses viral and host membranes through its S2 subunit. Two domains in S1 from different coronaviruses recognize a variety of host receptors, leading to viral attachment. The spike protein exists in two structurally distinct conformations, prefusion and postfusion. The transition from prefusion to postfusion conformation of the spike protein must be triggered, leading to membrane fusion. This article reviews current knowledge about the structures and functions of coronavirus spike proteins, illustrating how the two S1 domains recognize different receptors and how the spike proteins are regulated to undergo conformational transitions. I further discuss the evolution of these two critical functions of coronavirus spike proteins, receptor recognition and membrane fusion, in the context of the corresponding functions from other viruses and host cells.
Collapse
Affiliation(s)
- Fang Li
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota 55455;
| |
Collapse
|
38
|
Sinnott JA, Peisch SF, Tyekucheva S, Gerke T, Lis R, Rider JR, Fiorentino M, Stampfer MJ, Mucci LA, Loda M, Penney KL. Prognostic Utility of a New mRNA Expression Signature of Gleason Score. Clin Cancer Res 2016; 23:81-87. [PMID: 27663590 DOI: 10.1158/1078-0432.ccr-16-1245] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 07/27/2016] [Accepted: 09/15/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Gleason score strongly predicts prostate cancer mortality; however, scoring varies among pathologists, and many men are diagnosed with intermediate-risk Gleason score 7. We previously developed a 157-gene signature for Gleason score using a limited gene panel. Using a new whole-transcriptome expression dataset, we verified the previous signature's performance and developed a new Gleason signature to improve lethal outcome prediction among men with Gleason score 7. EXPERIMENTAL DESIGN We generated mRNA expression data from prostate tumor tissue from men in the Physicians' Health Study and Health Professionals Follow-Up Study (N = 404) using the Affymetrix Human Gene 1.0 ST microarray. The Prediction Analysis for Microarrays method was used to develop a signature to distinguish high (≥8) versus low (≤6) Gleason score. We evaluated the signature's ability to improve prediction of lethality among men with Gleason score 7, adjusting for 3 + 4/4 + 3 status, by quantifying the area under the receiver operating characteristic (ROC) curve (AUC). RESULTS We identified a 30-gene signature that best distinguished Gleason score ≤6 from ≥8. The AUC to predict lethal disease among Gleason score 7 men was 0.76 [95% confidence interval (CI), 0.67-0.84] compared with 0.68 (95% CI, 0.59-0.76) using 3 + 4/4 + 3 status alone (P = 0.0001). This signature was a nonsignificant (P = 0.09) improvement over our previous signature (AUC = 0.72). CONCLUSIONS Our new 30-gene signature improved prediction of lethality among men with Gleason score 7. This signature can potentially become a useful prognostic tool for physicians to improve treatment decision making. Clin Cancer Res; 23(1); 81-87. ©2016 AACRSee related commentary by Yin et al., p. 6.
Collapse
Affiliation(s)
- Jennifer A Sinnott
- Department of Statistics, Ohio State University, Columbus, Ohio.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Sam F Peisch
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Svitlana Tyekucheva
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Departments of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Travis Gerke
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida
| | - Rosina Lis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Jennifer R Rider
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts
| | | | - Meir J Stampfer
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Lorelei A Mucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Massimo Loda
- Pathology Unit, Addarii Institute, S. Orsola-Malpighi Hospital, Bologna, Italy.,Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Kathryn L Penney
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts. .,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
39
|
Wesley UV, Hatcher JF, Ayvaci ER, Klemp A, Dempsey RJ. Regulation of Dipeptidyl Peptidase IV in the Post-stroke Rat Brain and In Vitro Ischemia: Implications for Chemokine-Mediated Neural Progenitor Cell Migration and Angiogenesis. Mol Neurobiol 2016; 54:4973-4985. [PMID: 27525674 DOI: 10.1007/s12035-016-0039-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 08/05/2016] [Indexed: 12/13/2022]
Abstract
Cerebral ischemia evokes abnormal release of proteases in the brain microenvironment that spatiotemporally impact angio-neurogenesis. Dipeptidyl peptidase IV (DPPIV), a cell surface and secreted protease, has been implicated in extracellular matrix remodeling by regulating cell adhesion, migration, and angiogenesis through modifying the functions of the major chemokine stromal-derived factor, SDF1. To elucidate the possible association of DPPIV in ischemic brain, we examined the expression of DPPIV in the post-stroke rat brain and under in vitro ischemia by oxygen glucose deprivation (OGD). We further investigated the effects of DPPIV on SDF1 mediated in vitro chemotactic and angiogenic functions. DPPIV protein and mRNA levels were significantly upregulated during repair phase in the ischemic cortex of the rat brain, specifically in neurons, astrocytes, and endothelial cells. In vitro exposure of Neuro-2a neuronal cells and rat brain endothelial cells to OGD resulted in upregulation of DPPIV. In vitro functional analysis showed that DPPIV decreases the SDF1-mediated angiogenic potential of rat brain endothelial cells and inhibits the migration of Neuro-2a and neural progenitor cells. Western blot analyses revealed decreased levels of phosphorylated ERK1/2 and AKT in the presence of DPPIV. DPPIV inhibitor restored the effects of SDF1. Proteome profile array screening further revealed that DPPIV decreases matrix metalloproteinase-9, a key downstream effector of ERK-AKT signaling pathways. Overall, delayed induction of DPPIV in response to ischemia/reperfusion suggests that DPPIV may play an important role in endogenous brain tissue remodeling and repair processes. This may be mediated through modulation of SDF1-mediated cell migration and angiogenesis.
Collapse
Affiliation(s)
- Umadevi V Wesley
- Department of Neurological Surgery, University of Wisconsin, Clinical Science Center, 600 Highland Ave, Box 8660, Madison, WI, 53792, USA.
| | - James F Hatcher
- Department of Neurological Surgery, University of Wisconsin, Clinical Science Center, 600 Highland Ave, Box 8660, Madison, WI, 53792, USA
| | - Emine R Ayvaci
- Department of Neurological Surgery, University of Wisconsin, Clinical Science Center, 600 Highland Ave, Box 8660, Madison, WI, 53792, USA
| | - Abby Klemp
- Department of Neurological Surgery, University of Wisconsin, Clinical Science Center, 600 Highland Ave, Box 8660, Madison, WI, 53792, USA
| | - Robert J Dempsey
- Department of Neurological Surgery, University of Wisconsin, Clinical Science Center, 600 Highland Ave, Box 8660, Madison, WI, 53792, USA.
| |
Collapse
|
40
|
Khaket TP, Dhanda S, Jodha D, Singh J. Biochemical studies on dipeptidyl peptidase I (cathepsin C) from germinated Vigna radiata seeds. Process Biochem 2016. [DOI: 10.1016/j.procbio.2016.04.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
41
|
Gong Q, Shi W, Li L, Wu X, Ma H. Ultrasensitive Fluorescent Probes Reveal an Adverse Action of Dipeptide Peptidase IV and Fibroblast Activation Protein during Proliferation of Cancer Cells. Anal Chem 2016; 88:8309-14. [PMID: 27444320 DOI: 10.1021/acs.analchem.6b02231] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Dipeptide peptidase IV (DPPIV) and fibroblast activation protein (FAP) are isoenzymes. Evidence shows that DPPIV is related to antitumor immunity, and FAP may be a drug target in cancer therapy, making it seem that the two enzymes might have a synergistic role during the proliferation of cancer cells. Surprisingly, herein, we find an adverse action of DPPIV and FAP in the proliferation process by analyzing their changes with two tailor-made ultrasensitive fluorescent probes. First, the up-regulation of DPPIV and down-regulation of FAP in cancer cells under the stimulation of genistein are detected. Then, we find that MGC803 cells with a higher FAP but lower DPPIV level than SGC7901 cells exhibit a faster proliferation rate. Importantly, inhibiting the DPPIV expression with siRNA increases the proliferation rate of MGC803 cells, whereas the FAP inhibition decreases the rate. These findings suggest that the two enzymes play an adverse role during the proliferation of cancer cells, which provides us a new viewpoint for cancer studies.
Collapse
Affiliation(s)
- Qiuyu Gong
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190, China.,University of Chinese Academy of Sciences , Beijing 100049, China
| | - Wen Shi
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190, China
| | - Lihong Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190, China
| | - Xiaofeng Wu
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190, China
| | - Huimin Ma
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190, China.,University of Chinese Academy of Sciences , Beijing 100049, China
| |
Collapse
|
42
|
Xinhua W, Xiangting Q, Lingling C, Guohong L. Prognostic Significance of Serum CD26 Concentration in Patients with Esophageal Squamous Cell Carcinoma. Arch Med Res 2016; 47:299-303. [DOI: 10.1016/j.arcmed.2016.07.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 07/06/2016] [Indexed: 12/12/2022]
|
43
|
Beckenkamp A, Davies S, Willig JB, Buffon A. DPPIV/CD26: a tumor suppressor or a marker of malignancy? Tumour Biol 2016; 37:7059-73. [DOI: 10.1007/s13277-016-5005-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/25/2016] [Indexed: 12/12/2022] Open
|
44
|
Zhao W, Mazar J, Lee B, Sawada J, Li JL, Shelley J, Govindarajan S, Towler D, Mattick JS, Komatsu M, Dinger ME, Perera RJ. The Long Noncoding RNA SPRIGHTLY Regulates Cell Proliferation in Primary Human Melanocytes. J Invest Dermatol 2016; 136:819-828. [PMID: 26829028 DOI: 10.1016/j.jid.2016.01.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 12/14/2015] [Accepted: 12/14/2015] [Indexed: 01/29/2023]
Abstract
The long noncoding RNA SPRIGHTLY (formerly SPRY4-IT1), which lies within the intronic region of the SPRY4 gene, is up-regulated in human melanoma cells compared to melanocytes. SPRIGHTLY regulates a number of cancer hallmarks, including proliferation, motility, and apoptosis. To better understand its oncogenic role, SPRIGHTLY was stably transfected into human melanocytes, which resulted in increased cellular proliferation, colony formation, invasion, and development of a multinucleated dendritic-like phenotype. RNA sequencing and mass spectrometric analysis of SPRIGHTLY-expressing cells revealed changes in the expression of genes involved in cell proliferation, apoptosis, chromosome organization, regulation of DNA damage responses, and cell cycle. The proliferation marker Ki67, minichromosome maintenance genes 2-5, antiapoptotic gene X-linked inhibitor of apoptosis, and baculoviral IAP repeat-containing 7 were all up-regulated in SPRIGHTLY-expressing melanocytes, whereas the proapoptotic tumor suppressor gene DPPIV/CD26 was down-regulated, followed by an increase in extracellular signal-regulated kinase 1/2 phosphorylation, suggesting an increase in mitogen-activated protein kinase activity. Because down-regulation of DPPIV is known to be associated with malignant transformation in melanocytes, SPRIGHTLY-mediated DPPIV down-regulation may play an important role in melanoma pathobiology. Together, these findings provide important insights into how SPRIGHTLY regulates cell proliferation and anchorage-independent colony formation in primary human melanocytes.
Collapse
Affiliation(s)
- Wei Zhao
- Sanford-Burnham Medical Research Institute, Orlando, Florida, USA
| | - Joseph Mazar
- Sanford-Burnham Medical Research Institute, Orlando, Florida, USA
| | - Bongyong Lee
- Sanford-Burnham Medical Research Institute, Orlando, Florida, USA
| | - Junko Sawada
- Sanford-Burnham Medical Research Institute, Orlando, Florida, USA
| | - Jian-Liang Li
- Sanford-Burnham Medical Research Institute, Orlando, Florida, USA
| | - John Shelley
- Sanford-Burnham Medical Research Institute, Orlando, Florida, USA
| | | | - Dwight Towler
- Sanford-Burnham Medical Research Institute, Orlando, Florida, USA
| | - John S Mattick
- Garvan Institute of Medical Research and St. Vincent's Clinical School, University of New South Wales, Darlinghurst, Australia
| | - Masanobu Komatsu
- Sanford-Burnham Medical Research Institute, Orlando, Florida, USA
| | - Marcel E Dinger
- Garvan Institute of Medical Research and St. Vincent's Clinical School, University of New South Wales, Darlinghurst, Australia
| | - Ranjan J Perera
- Sanford-Burnham Medical Research Institute, Orlando, Florida, USA.
| |
Collapse
|
45
|
Li X, Wang C, Xiao J, McKeehan WL, Wang F. Fibroblast growth factors, old kids on the new block. Semin Cell Dev Biol 2016; 53:155-67. [PMID: 26768548 DOI: 10.1016/j.semcdb.2015.12.014] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 12/18/2015] [Indexed: 01/08/2023]
Abstract
The fibroblast growth factors (FGFs) are a family of cell intrinsic regulatory peptides that control a broad spectrum of cellular activities. The family includes canonic FGFs that elicit their activities by activating the FGF receptor (FGFR) tyrosine kinase and non-canonic members that elicit their activities intracellularly and via FGFR-independent mechanisms. The FGF signaling axis is highly complex due to the existence of multiple isoforms of both ligands and receptors, as well as cofactors that include the chemically heterogeneous heparan sulfate (HS) cofactors, and in the case of endocrine FGFs, the Klotho coreceptors. Resident FGF signaling controls embryonic development, maintains tissue homeostasis, promotes wound healing and tissue regeneration, and regulates functions of multiple organs. However, ectopic or aberrant FGF signaling is a culprit for various diseases, including congenital birth defects, metabolic disorder, and cancer. The molecular mechanisms by which the specificity of FGF signaling is achieved remain incompletely understood. Since its application as a druggable target has been gradually recognized by pharmaceutical companies and translational researchers, understanding the determinants of FGF signaling specificity has become even more important in order to get into the position to selectively suppress a particular pathway without affecting others to minimize side effects.
Collapse
Affiliation(s)
- Xiaokun Li
- College of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cong Wang
- College of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Jian Xiao
- College of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wallace L McKeehan
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030-3303, United States
| | - Fen Wang
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030-3303, United States.
| |
Collapse
|
46
|
Feng M, Fang Y, Han B, Xu X, Fan P, Hao Y, Qi Y, Hu H, Huo X, Meng L, Wu B, Li J. In-Depth N-Glycosylation Reveals Species-Specific Modifications and Functions of the Royal Jelly Protein from Western (Apis mellifera) and Eastern Honeybees (Apis cerana). J Proteome Res 2015; 14:5327-40. [PMID: 26496797 DOI: 10.1021/acs.jproteome.5b00829] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Royal jelly (RJ), secreted by honeybee workers, plays diverse roles as nutrients and defense agents for honeybee biology and human health. Despite being reported to be glycoproteins, the glycosylation characterization and functionality of RJ proteins in different honeybee species are largely unknown. An in-depth N-glycoproteome analysis and functional assay of RJ produced by Apis mellifera lingustica (Aml) and Apis cerana cerana (Acc) were conducted. RJ produced by Aml yielded 80 nonredundant N-glycoproteins carrying 190 glycosites, of which 23 novel proteins harboring 35 glycosites were identified. For Acc, all 43 proteins glycosylated at 138 glycosites were reported for the first time. Proteins with distinct N-glycoproteomic characteristics in terms of glycoprotein species, number of N-glycosylated sites, glycosylation motif, abundance level of glycoproteins, and N-glycosites were observed in this two RJ samples. The fact that the low inhibitory efficiency of N-glycosylated major royal jelly protein 2 (MRJP2) against Paenibacillus larvae (P. larvae) and the absence of antibacterial related glycosylated apidaecin, hymenoptaecin, and peritrophic matrix in the Aml RJ compared to Acc reveal the mechanism for why the Aml larvae are susceptible to P. larvae, the causative agent of a fatal brood disease (American foulbrood, AFB). The observed antihypertension activity of N-glycosylated MRJP1 in two RJ samples and a stronger activity found in Acc than in Aml reveal that specific RJ protein and modification are potentially useful for the treatment of hypertensive disease for humans. Our data gain novel understanding that the western and eastern bees have evolved species-specific strategies of glycosylation to fine-tune protein activity for optimizing molecular function as nutrients and immune agents for the good of honeybee and influence on the health promoting activity for human as well. This serves as a valuable resource for the targeted probing of the biological functions of RJ proteins for honeybee and medical communities.
Collapse
Affiliation(s)
- Mao Feng
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences , Beijing 100093, China
| | - Yu Fang
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences , Beijing 100093, China
| | - Bin Han
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences , Beijing 100093, China
| | - Xiang Xu
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences , Beijing 100093, China
| | - Pei Fan
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences , Beijing 100093, China.,College of Bioengineering, Henan University of Technology , Zhengzhou 450001, China
| | - Yue Hao
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences , Beijing 100093, China
| | - Yuping Qi
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences , Beijing 100093, China
| | - Han Hu
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences , Beijing 100093, China
| | - Xinmei Huo
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences , Beijing 100093, China
| | - Lifeng Meng
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences , Beijing 100093, China
| | - Bin Wu
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences , Beijing 100093, China
| | - Jianke Li
- Institute of Apicultural Research/Key Laboratory of Pollinating Insect Biology, Ministry of Agriculture, Chinese Academy of Agricultural Sciences , Beijing 100093, China
| |
Collapse
|
47
|
Boccardi V, Marano L, Rossetti RRA, Rizzo MR, di Martino N, Paolisso G. Serum CD26 levels in patients with gastric cancer: a novel potential diagnostic marker. BMC Cancer 2015; 15:703. [PMID: 26471376 PMCID: PMC4608357 DOI: 10.1186/s12885-015-1757-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Accepted: 10/09/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND CD26 is an ectoenzyme with dipeptidyl peptidase 4 (DPP4) activity expressed on a variety of cell types. Considering that serum CD26 levels have been previously associated with different cancers, we examined the potential diagnostic value of serum CD26 levels in gastric cancer. METHODS Soluble serum CD26 levels were measured in pre and postoperative serum samples of 30 patients with gastric cancer and in 24 healthy donors by a specific ELISA kit. RESULTS We found significantly lower serum CD26 levels in patients with gastric cancer (557.7 ± 118.3 pg/mL) compared with healthy donors (703.4 ± 170.3 pg/mL). Moreover patients with HER2 positive tumors had significantly lower CD26 serum levels (511.8 ± 84.8 pg/mL) compared with HER2 negative tumors (619.1 ± 109.9 pg/mL, p = 0.006). A binary logistic model having gastric cancer as the dependent variable while age, gender, CEA, CA19.9 and CD26 levels as covariates, showed that CD26 serum levels were independently associated with gastric cancer presence. Indeed after 3 months from surgery serum CD26 levels significantly increased (700.1 ± 119.9 pg/mL vs 557.7 ± 118.3 pg/ml) in all patients (t = -4.454, p < 0.0001). CONCLUSIONS This is a preliminary study showing that the measurement of serum CD26 levels could represent an early detection marker for gastric cancer.
Collapse
Affiliation(s)
- Virginia Boccardi
- Department of Internal Medicine, Surgical, Neurological Metabolic Disease and Geriatric Medicine, Second University of Naples, Piazza Miraglia 2, 80138, Naples, Italy.
| | - Luigi Marano
- General, Minimally Invasive and Robotic Surgery, Department of Surgery, "San Matteo degli Infermi" Hospital, ASL Umbria 2, 06049, Spoleto PG, Italy.
| | - Rosaria Rita Amalia Rossetti
- Department of Internal Medicine, Surgical, Neurological Metabolic Disease and Geriatric Medicine, Second University of Naples, Piazza Miraglia 2, 80138, Naples, Italy.
| | - Maria Rosaria Rizzo
- Department of Internal Medicine, Surgical, Neurological Metabolic Disease and Geriatric Medicine, Second University of Naples, Piazza Miraglia 2, 80138, Naples, Italy.
| | - Natale di Martino
- Department of Internal Medicine, Surgical, Neurological Metabolic Disease and Geriatric Medicine, Second University of Naples, Piazza Miraglia 2, 80138, Naples, Italy.
| | - Giuseppe Paolisso
- Department of Internal Medicine, Surgical, Neurological Metabolic Disease and Geriatric Medicine, Second University of Naples, Piazza Miraglia 2, 80138, Naples, Italy.
| |
Collapse
|
48
|
Prognostic relevance of stromal CD26 expression in rectal cancer after chemoradiotherapy. Int J Clin Oncol 2015; 21:350-358. [PMID: 26370256 DOI: 10.1007/s10147-015-0902-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 08/30/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND CD26 is a transmembrane glycoprotein whose role in various types of malignancies, along with the potential therapeutic and diagnostic targets, has been evaluated. Preoperative chemoradiotherapy (CRT) is an effective tool for local control of rectal cancer, but the rate of disease recurrence remains high. The aim of this study was to clarify the association between CD26 expression and rectal cancer after preoperative CRT. METHODS A total of 85 patients with rectal cancer who had undergone preoperative CRT were enrolled in this study. We investigated CD26 expression in residual tumors and the surrounding stromal tissue using immunohistochemistry. Additionally, stromal CD26 gene expression was assessed by real-time quantitative polymerase chain reaction. RESULTS Patients with high CD26 expression in cancer tissue more frequently had serosal invasion, vascular invasion, and a poor pathological response. High expression of CD26 in the tumor stroma was significantly correlated with histology and tumor recurrence. High CD26 expression in the stroma, but not the tumor itself, was significantly correlated with a poor prognosis. Patients expressing CD26 in the tumor stroma, based on transcriptional analysis, also had a significantly poorer prognosis than those without the expression. In multivariate analysis, lymph node metastasis and high stromal CD26 expression were identified as independent prognostic factors in patients with rectal cancer after neoadjuvant CRT. CONCLUSION Stromal CD26 expression after preoperative CRT was significantly associated with tumor recurrence and prognosis in rectal cancer patients. Our data suggest that stromal CD26 plays an important role and is a potential therapeutic target in tumor relapse.
Collapse
|
49
|
Beckenkamp A, Willig JB, Santana DB, Nascimento J, Paccez JD, Zerbini LF, Bruno AN, Pilger DA, Wink MR, Buffon A. Differential Expression and Enzymatic Activity of DPPIV/CD26 Affects Migration Ability of Cervical Carcinoma Cells. PLoS One 2015. [PMID: 26222679 PMCID: PMC4519168 DOI: 10.1371/journal.pone.0134305] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Dipeptidyl peptidase IV (DPPIV/CD26) is a transmembrane glycoprotein that inactivates or degrades some bioactive peptides and chemokines. For this reason, it regulates cell proliferation, migration and adhesion, showing its role in cancer processes. This enzyme is found mainly anchored onto the cell membrane, although it also has a soluble form, an enzymatically active isoform. In the present study, we investigated DPPIV/CD26 activity and expression in cervical cancer cell lines (SiHa, HeLa and C33A) and non-tumorigenic HaCaT cells. The effect of the DPPIV/CD26 inhibitor (sitagliptin phosphate) on cell migration and adhesion was also evaluated. Cervical cancer cells and keratinocytes exhibited DPPIV/CD26 enzymatic activity both membrane-bound and in soluble form. DPPIV/CD26 expression was observed in HaCaT, SiHa and C33A, while in HeLa cells it was almost undetectable. We observed higher migratory capacity of HeLa, when compared to SiHa. But in the presence of sitagliptin SiHa showed an increase in migration, indicating that, at least in part, cell migration is regulated by DPPIV/CD26 activity. Furthermore, in the presence of sitagliptin phosphate, SiHa and HeLa cells exhibited a significant reduction in adhesion. However this mechanism seems to be mediated independent of DPPIV/CD26. This study demonstrates, for the first time, the activity and expression of DPPIV/CD26 in cervical cancer cells and the effect of sitagliptin phosphate on cell migration and adhesion.
Collapse
Affiliation(s)
- Aline Beckenkamp
- Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Júlia Biz Willig
- Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Danielle Bertodo Santana
- Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Jéssica Nascimento
- Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Juliano Domiraci Paccez
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cancer Genomics Group, Cape Town, South Africa
| | - Luiz Fernando Zerbini
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cancer Genomics Group, Cape Town, South Africa
| | | | - Diogo André Pilger
- Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Márcia Rosângela Wink
- Laboratory of Cell Biology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS, Brazil
| | - Andréia Buffon
- Pharmaceutical Sciences Graduate Program, Faculty of Pharmacy, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
- * E-mail:
| |
Collapse
|
50
|
Matyszewski A, Czarnecka A, Kawecki M, Korzeń P, Safir IJ, Kukwa W, Szczylik C. Impaired glucose metabolism treatment and carcinogenesis. Oncol Lett 2015; 10:589-594. [PMID: 26622538 DOI: 10.3892/ol.2015.3324] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 04/29/2015] [Indexed: 12/29/2022] Open
Abstract
Carbohydrate metabolism disorders increase the risk of carcinogenesis. Diabetes mellitus alters numerous physiological processes that may encourage cancer growth. However, treating impaired glucose homeostasis may actually promote neoplasia; maintaining proper glucose plasma concentrations reduces metabolic stresses, however, certain medications may themselves result in oncogenic effects. A number of previous studies have demonstrated that metformin reduces the cancer risk. However, the use of sulfonylurea derivatives correlates with an increased risk of developing a malignancy. Another form of treatment, insulin therapy, involves using various forms of insulin that differ in pharmacodynamics, pharmacokinetics and efficacy. Previous studies have indicated that certain insulin variants also affect the cancer risk. The results from analyses that address the safety of long-lasting insulin types raise the most concern regarding the increased risk of malignancy. Rapid development of novel diabetic medications and their widespread use carries the risk of potentially increased rates of cancer, unnoticeable in limited, randomized, controlled trials. In the present review, the results of clinical and epidemiological studies are evaluated to assess the safety of anti-hyperglycemic medications and their effect on cancer risk and outcomes.
Collapse
Affiliation(s)
- Artur Matyszewski
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland ; Department of Internal Medicine, Cardiology and Hypertensionr, L. Rydygier Province Hospital, Suwałki, Poland ; Department of Nephrology and Dialysis Center, L. Rydygier Province Hospital, Suwałki, Poland
| | - Anna Czarnecka
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Maciej Kawecki
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Piotr Korzeń
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Ilan J Safir
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Wojciech Kukwa
- Department of Otorhinolaryngology, Medical University of Warsaw, Warsaw, Poland
| | - Cezary Szczylik
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| |
Collapse
|