1
|
Sun L, Ma Y, Geng C, Gao X, Li X, Ru Q, Zhu S, Zhang P. DPP4, a potential tumor biomarker, and tumor therapeutic target: review. Mol Biol Rep 2025; 52:126. [PMID: 39821530 DOI: 10.1007/s11033-025-10235-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025]
Abstract
Dipeptidyl peptidase 4 (DPP4) is a serine protease widely distributed in membrane-bound and soluble forms in various tissues and organs throughout the body. DPP4 plays a role in inflammation, immune regulation, cell growth, migration and differentiation. The role of DPP4 in tumors has garnered increasing attention. Previous research has demonstrated that DPP4 contributes to the promotion of cancer in most cancers, and it may play a specific biological function through the variation in tumor cell types and expression forms. However, the expression of DDP4 in different tumor types and its specific mechanism remains unclear. In this review, we describe the structure of DPP4, summarize the recent research progress of its expression and potential mechanisms in common tumors, and discuss the development prospects of DPP4 inhibitors in tumor therapy. Although current research emphasizes the potential of DPP4 as a drug target, the incomplete understanding of its regulatory mechanisms impedes the discovery and development of new therapies against it. Further research on DPP4-related tumors is anticipated to promote its clinical application as a potential therapeutic target.
Collapse
Affiliation(s)
- Lu Sun
- Department of Ultrasound, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266035, China
| | - Yuhui Ma
- Department of Ultrasound, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266035, China
| | - Chenchen Geng
- Department of Ultrasound, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266035, China
| | - Xiaoqian Gao
- Department of Ultrasound, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266035, China
| | - Xinbing Li
- Department of Ultrasound, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266035, China
| | - Qi Ru
- Department of Ultrasound, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266035, China
| | - Shuzhen Zhu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China.
- Department of Clinical Laboratory, Qilu Hospital of Shandong University (Qingdao), Qingdao, Shandong, 266035, China.
| | - Ping Zhang
- Department of Ultrasound, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266035, China.
- Health Management Center, Qilu Hospital of Shandong University (Qingdao), Qingdao, 266035, China.
| |
Collapse
|
2
|
Chellappan MC, Vasu S, Mahadevan S, Kathiravan MK, Jayaraman S, Naik S. Beneficial Effects of PIN1 Inhibition on Diabetes Mellitus: A Concise Review. Endocr Metab Immune Disord Drug Targets 2025; 25:2-7. [PMID: 38693739 DOI: 10.2174/0118715303297663240307060019] [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: 01/20/2024] [Revised: 02/20/2024] [Accepted: 02/26/2024] [Indexed: 05/03/2024]
Abstract
Type 2 diabetes mellitus is a long-term medical illness in which the body either becomes resistant to insulin or fails to produce it sufficiently. Mostly, combinatorial therapy is required to control blood glucose levels. However, combinatorial therapy has detrimental side effects. The prevalence of the cases and subsequent increases in medical costs of the same intimidate human health globally. While there have been a lot of studies focused on developing diabetic regimens that work to lower blood glucose levels, their effectiveness is short-lived because of unfavorable side effects, such as weight gain and hypoglycemia. In recent years, the PIN1 (protein interacting with NIMA) enzyme has attracted the attention of researchers. Previous studies suggested that PIN1 may act on the various substrates that are involved in the progression of T2DM and also help in the management of diabetes-related disorders. Thus, the focus of the current review is to examine the correlation between PIN1, T2DM and its related disorders and explore the possibility of developing novel therapeutic targets through PIN1 inhibition.
Collapse
Affiliation(s)
- Meeramol C Chellappan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, (DU) Porur-600 116, Chennai, India
| | - Soumya Vasu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, (DU) Porur-600 116, Chennai, India
| | - Shriraam Mahadevan
- Department of Endocrinology, Sri Ramachandra Institute of Higher Education and Research, (DU) Porur-600 116, Chennai, India
| | | | - Saravanan Jayaraman
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, 643001, India
| | - Soniya Naik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sri Ramachandra Institute of Higher Education and Research, (DU) Porur-600 116, Chennai, India
| |
Collapse
|
3
|
Niazmand A, Nedaeinia R, Vatandoost N, Jafarpour S, Safabakhsh S, Kolahdouz M, Ferns GA, Salehi R. The impacts of dipeptidyl- peptidase 4 (DPP-4) inhibitors on common female malignancies: A systematic review. Gene 2024; 927:148659. [PMID: 38866262 DOI: 10.1016/j.gene.2024.148659] [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: 09/25/2023] [Revised: 05/29/2024] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
The inhibition of dipeptidyl- peptidase 4 (DPP-4) is an essential therapy for controlling hyperglycemia in patients with type 2 diabetes (T2DM). However, the role of DPP-4 in cancer is not yet clear, with some studies suggesting that it may either promote or suppress tumors. This makes it crucial to have personalized treatment for diabetic women with cancer to effectively manage their diabetes whilst and preventing cancer mortality. To address this issue, we conducted an integrative in-silico analysis and systematic review of the literature to comprehensively examine the relationship between DPP-4 expression and the effects of its inhibitors on prevalent female malignancies. We specifically chose studies that examined the effects of DPP-4 expression and DPP-4 inhibition (DPP-4i) on prevalent cancers in women, such as breast cancer (BC), ovarian cancer (OV), cervical cancer (CC), and endometrial cancer (EC). These studies comprised those conducted both in vivo and in vitro. The review of the literature indicated that DPP-4i may worsen aggressive traits such as metastasis, Epithelial-to-mesenchymal transition (EMT), and chemotherapy resistance in BC cells. However, cohort studies on diabetic and BC patients did not confirm these findings. In vitro studies indicate that on OV, DPP-4 upregulation has been shown to prevent metastasis, while CCappears to be influenced by DPP-4 expression in terms of cell migration. sitagliptin, a pharmaceutical inhibitor of DPP-4, had a significant impact on reducing adhesion in CC cells in vitro. Overexpression of DPP-4 increased cell migration and proliferation in CC and EC cells, and hence the application of sitagliptin is expected to prevent this effect. On the other hand, the result of in-silico data confirmed that a significant correlation exists between DPP-4 expression and immune cell infiltration in breast, ovarian, cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC) as well as downregulated in these cancers compared to their normal tissue samples. Furthermore, a significant (p < 0.05) effect on OS of BC and CESC patients has been reported due to the elevation of DPP-4 methylation on a specific CPG Island. These findings could aid in creating specialized treatments for diabetic women with specific malignancies, but caution should be exercised when considering the patient's medical history and cancer type.
Collapse
Affiliation(s)
- Anoosha Niazmand
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nasimeh Vatandoost
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sima Jafarpour
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saeid Safabakhsh
- Micronesian Institute for Disease Prevention and Research, 736 Route 4, Suite 103, Sinajana, GU 96910, USA
| | - Mahsa Kolahdouz
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Falmer, Brighton BN1 9PH, Sussex, UK
| | - Rasoul Salehi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
4
|
Dhas Y, Biswas N, M R D, Jones LD, Ashili S. Repurposing metabolic regulators: antidiabetic drugs as anticancer agents. MOLECULAR BIOMEDICINE 2024; 5:40. [PMID: 39333445 PMCID: PMC11436690 DOI: 10.1186/s43556-024-00204-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/26/2024] [Indexed: 09/29/2024] Open
Abstract
Drug repurposing in cancer taps into the capabilities of existing drugs, initially designed for other ailments, as potential cancer treatments. It offers several advantages over traditional drug discovery, including reduced costs, reduced development timelines, and a lower risk of adverse effects. However, not all drug classes align seamlessly with a patient's condition or long-term usage. Hence, repurposing of chronically used drugs presents a more attractive option. On the other hand, metabolic reprogramming being an important hallmark of cancer paves the metabolic regulators as possible cancer therapeutics. This review emphasizes the importance and offers current insights into the repurposing of antidiabetic drugs, including metformin, sulfonylureas, sodium-glucose cotransporter 2 (SGLT2) inhibitors, dipeptidyl peptidase 4 (DPP-4) inhibitors, glucagon-like peptide-1 receptor agonists (GLP-1RAs), thiazolidinediones (TZD), and α-glucosidase inhibitors, against various types of cancers. Antidiabetic drugs, regulating metabolic pathways have gained considerable attention in cancer research. The literature reveals a complex relationship between antidiabetic drugs and cancer risk. Among the antidiabetic drugs, metformin may possess anti-cancer properties, potentially reducing cancer cell proliferation, inducing apoptosis, and enhancing cancer cell sensitivity to chemotherapy. However, other antidiabetic drugs have revealed heterogeneous responses. Sulfonylureas and TZDs have not demonstrated consistent anti-cancer activity, while SGLT2 inhibitors and DPP-4 inhibitors have shown some potential benefits. GLP-1RAs have raised concerns due to possible associations with an increased risk of certain cancers. This review highlights that further research is warranted to elucidate the mechanisms underlying the potential anti-cancer effects of these drugs and to establish their efficacy and safety in clinical settings.
Collapse
Affiliation(s)
- Yogita Dhas
- Rhenix Lifesciences, Hyderabad, 500038, Telangana, India
| | - Nupur Biswas
- Rhenix Lifesciences, Hyderabad, 500038, Telangana, India.
- CureScience, 5820 Oberlin Dr, Suite 202, San Diego, CA, 92121, USA.
| | | | - Lawrence D Jones
- CureScience, 5820 Oberlin Dr, Suite 202, San Diego, CA, 92121, USA
| | | |
Collapse
|
5
|
Tóth F, Moftakhar Z, Sotgia F, Lisanti MP. In Vitro Investigation of Therapy-Induced Senescence and Senescence Escape in Breast Cancer Cells Using Novel Flow Cytometry-Based Methods. Cells 2024; 13:841. [PMID: 38786063 PMCID: PMC11120107 DOI: 10.3390/cells13100841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Although cellular senescence was originally defined as an irreversible form of cell cycle arrest, in therapy-induced senescence models, the emergence of proliferative senescence-escaped cancer cells has been reported by several groups, challenging the definition of senescence. Indeed, senescence-escaped cancer cells may contribute to resistance to cancer treatment. Here, to study senescence escape and isolate senescence-escaped cells, we developed novel flow cytometry-based methods using the proliferation marker Ki-67 and CellTrace CFSE live-staining. We investigated the role of a novel senescence marker (DPP4/CD26) and a senolytic drug (azithromycin) on the senescence-escaping ability of MCF-7 and MDA-MB-231 breast cancer cells. Our results show that the expression of DPP4/CD26 is significantly increased in both senescent MCF-7 and MDA-MB-231 cells. While not essential for senescence induction, DPP4/CD26 contributed to promoting senescence escape in MCF-7 cells but not in MDA-MB-231 cells. Our results also confirmed the potential senolytic effect of azithromycin in senescent cancer cells. Importantly, the combination of azithromycin and a DPP4 inhibitor (sitagliptin) demonstrated a synergistic effect in senescent MCF-7 cells and reduced the number of senescence-escaped cells. Although further research is needed, our results and novel methods could contribute to the investigation of the mechanisms of senescence escape and the identification of potential therapeutic targets. Indeed, DPP4/CD26 could be a promising marker and a novel target to potentially decrease senescence escape in cancer.
Collapse
Affiliation(s)
- Fanni Tóth
- Translational Medicine, University of Salford, Salford M5 4WT, UK; (F.T.)
- The CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT 25.3, 1090 Wien, Vienna, Austria
| | - Zahra Moftakhar
- Translational Medicine, University of Salford, Salford M5 4WT, UK; (F.T.)
| | - Federica Sotgia
- Translational Medicine, University of Salford, Salford M5 4WT, UK; (F.T.)
| | - Michael P. Lisanti
- Translational Medicine, University of Salford, Salford M5 4WT, UK; (F.T.)
| |
Collapse
|
6
|
Schwartz J, Capistrano KJ, Gluck J, Hezarkhani A, Naqvi AR. SARS-CoV-2, periodontal pathogens, and host factors: The trinity of oral post-acute sequelae of COVID-19. Rev Med Virol 2024; 34:e2543. [PMID: 38782605 PMCID: PMC11260190 DOI: 10.1002/rmv.2543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/04/2024] [Accepted: 05/09/2024] [Indexed: 05/25/2024]
Abstract
COVID-19 as a pan-epidemic is waning but there it is imperative to understand virus interaction with oral tissues and oral inflammatory diseases. We review periodontal disease (PD), a common inflammatory oral disease, as a driver of COVID-19 and oral post-acute-sequelae conditions (PASC). Oral PASC identifies with PD, loss of teeth, dysgeusia, xerostomia, sialolitis-sialolith, and mucositis. We contend that PD-associated oral microbial dysbiosis involving higher burden of periodontopathic bacteria provide an optimal microenvironment for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. These pathogens interact with oral epithelial cells activate molecular or biochemical pathways that promote viral adherence, entry, and persistence in the oral cavity. A repertoire of diverse molecules identifies this relationship including lipids, carbohydrates and enzymes. The S protein of SARS-CoV-2 binds to the ACE2 receptor and is activated by protease activity of host furin or TRMPSS2 that cleave S protein subunits to promote viral entry. However, PD pathogens provide additional enzymatic assistance mimicking furin and augment SARS-CoV-2 adherence by inducing viral entry receptors ACE2/TRMPSS, which are poorly expressed on oral epithelial cells. We discuss the mechanisms involving periodontopathogens and host factors that facilitate SARS-CoV-2 infection and immune resistance resulting in incomplete clearance and risk for 'long-haul' oral health issues characterising PASC. Finally, we suggest potential diagnostic markers and treatment avenues to mitigate oral PASC.
Collapse
Affiliation(s)
- Joel Schwartz
- Department of Oral Medicine and Diagnostic Sciences, University of Illinois Chicago, Chicago, Illinois, 60612, USA
| | | | - Joseph Gluck
- Department of Periodontics, University of Illinois Chicago, Chicago, Illinois, 60612, USA
| | - Armita Hezarkhani
- Department of Periodontics, University of Illinois Chicago, Chicago, Illinois, 60612, USA
| | - Afsar R. Naqvi
- Department of Periodontics, University of Illinois Chicago, Chicago, Illinois, 60612, USA
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, 60612, USA
| |
Collapse
|
7
|
Ma P, Ou H, Sun Z, Lu Y, Li M, Xu L, Liang Y, Zheng J, Ou Y. IAVPGEVA: Orally Available DPP4-Targeting Soy Glycinin Derived Octapeptide with Therapeutic Potential in Nonalcoholic Steatohepatitis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:7167-7178. [PMID: 38511978 DOI: 10.1021/acs.jafc.3c08932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
IAVPGEVA, an octapeptide derived from soybean 11S globulin hydrolysis, also known as SGP8, has exhibited regulatory effects on lipid metabolism, inflammation, and fibrosis in vitro. Studies using MCD and HFD-induced nonalcoholic steatohepatitis (NASH) models in mice show that SGP8 attenuates hepatic injury and metabolic disorders. Mechanistic studies suggest that SGP8 inhibits the JNK-c-Jun pathway in L02 cells and liver tissue under metabolic stress and targets DPP4 with DPP4 inhibitory activity. In conclusion, the results suggest that SGP8 is an orally available DPP4-targeting peptide with therapeutic potential in NASH.
Collapse
Affiliation(s)
- Peng Ma
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, Jiangsu, China
| | - Hao Ou
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, Jiangsu, China
| | - Zhongkan Sun
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, Jiangsu, China
| | - Yunbiao Lu
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, Jiangsu, China
| | - Mengdan Li
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, Jiangsu, China
| | - Liuxin Xu
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, Jiangsu, China
| | - Yan Liang
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, Jiangsu, China
| | - Jiawei Zheng
- School of Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, Jiangsu, China
| | - Yu Ou
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, Jiangsu, China
| |
Collapse
|
8
|
You F, Li C, Zhang S, Zhang Q, Hu Z, Wang Y, Zhang T, Meng Q, Yu R, Gao S. Sitagliptin inhibits the survival, stemness and autophagy of glioma cells, and enhances temozolomide cytotoxicity. Biomed Pharmacother 2023; 162:114555. [PMID: 36966667 DOI: 10.1016/j.biopha.2023.114555] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 03/10/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
The standard regimen treatment has improved GBM outcomes, but the survival rate of patients is still unsatisfactory. Temozolomide (TMZ) resistance is one of main reasons limiting the therapeutic efficacy of GBM. However, there are currently no TMZ-sensitizing drugs available in the clinic. Here we aimed to study whether the antidiabetic drug Sitagliptin can inhibit the survival, stemness and autophagy of GBM cells, and thus enhance TMZ cytotoxicity. We used CCK-8, EdU, colony formation, TUNEL and flow cytometry assays to assess cell proliferation and apoptosis; sphere formation and limiting dilution assays to measure self-renewal and stemness of glioma stem cells (GSCs); Western blot, qRT-PCR or immunohistochemical analysis to measure the expression of proliferation or stem cell markers; Western blot/fluorescent analysis of LC3 and other molecules to evaluate autophagy formation and degradation in glioma cells. We found that Sitagliptin inhibited proliferation and induced apoptosis in GBM cells and suppressed self-renewal and stemness of GSCs. The in vitro findings were further confirmed in glioma intracranial xenograft models. Sitagliptin administration prolonged the survival time of tumor-bearing mice. Sitagliptin could inhibit TMZ-induced protective autophagy and enhance the cytotoxicity of TMZ in glioma cells. In addition, Sitagliptin acted as a dipeptidyl peptidase 4 inhibitor in glioma as well as in diabetes, but it did not affect the blood glucose level and body weight of mice. These findings suggest that Sitagliptin with established pharmacologic and safety profiles could be repurposed as an antiglioma drug to overcome TMZ resistance, providing a new option for GBM therapy.
Collapse
|
9
|
Sun H, Qi X. The role of insulin and incretin-based drugs in biliary tract cancer: epidemiological and experimental evidence. Discov Oncol 2022; 13:70. [PMID: 35933633 PMCID: PMC9357599 DOI: 10.1007/s12672-022-00536-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 07/26/2022] [Indexed: 11/29/2022] Open
Abstract
Insulin and incretin-based drugs are important antidiabetic agents with complex effects on cell growth and metabolism. Emerging evidence shows that insulin and incretin-based drugs are associated with altered risk of biliary tract cancer (BTC). Observational study reveals that insulin is associated with an increased risk of extrahepatic cholangiocarcinoma (ECC), but not intrahepatic cholangiocarcinoma (ICC) or gallbladder cancer (GBC). This type-specific effect can be partly explained by the cell of origin and heterogeneous genome landscape of the three subtypes of BTC. Similar to insulin, incretin-based drugs also exhibit very interesting contradictions and inconsistencies in response to different cancer phenotypes, including BTC. Both epidemiological and experimental evidence suggests that incretin-based drugs can be a promoter of some cancers and an inhibitor of others. It is now more apparent that this type of drugs has a broader range of physiological effects on the body, including regulation of endoplasmic reticulum stress, autophagy, metabolic reprogramming, and gene expression. In particular, dipeptidyl peptidase-4 inhibitors (DPP-4i) have a more complex effect on cancer due to the multi-functional nature of DPP-4. DPP-4 exerts both catalytic and non-enzymatic functions to regulate metabolic homeostasis, immune reaction, cell migration, and proliferation. In this review, we collate the epidemiological and experimental evidence regarding the effect of these two classes of drugs on BTC to provide valuable information.
Collapse
Affiliation(s)
- Hua Sun
- Department of Geriatrics, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, No.208 East Huancheng Road, Hangzhou, Zhejiang, China
| | - Xiaohui Qi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.573 Xujiahui Road, Shanghai, China.
| |
Collapse
|
10
|
Salama MM, Zaghloul RA, Khalil RM, El-Shishtawy MM. Sitagliptin Potentiates the Anti-Neoplastic Activity of Doxorubicin in Experimentally-Induced Mammary Adenocarcinoma in Mice: Implication of Oxidative Stress, Inflammation, Angiogenesis, and Apoptosis. Sci Pharm 2022; 90:42. [DOI: 10.3390/scipharm90030042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022] Open
Abstract
Sitagliptin (STG) is a highly selective dipeptidyl peptidase-4 inhibitor recently used in the treatment of type 2 diabetes. The current study aimed to investigate the anti-neoplastic effect of STG alone and in combination with Doxorubicin (Dox), a known chemotherapeutic agent but with ominous side effects. After intramuscular inoculation of 2 × 106 Ehrlich tumor cells, Female Swiss mice were divided into tumor-bearing control, STG-treated, Dox-treated, and a combination of STG and Dox-treated groups. The results showed a significant reduction in the tumor growth of the treated animals in comparison with those of the positive control group with a more prominent effect in the co-treated group. Where, the anti-proliferative and apoptotic effect of STG, and its chemo-sensitizing ability, when used in combination with Dox, was mediated by modulation of oxidative stress (MDA and GSH), attenuation of tumor inflammation (IL-6 and IL-1β), and angiogenesis (VEGF), suppressing proliferation (β-catenin and cyclin-D1) and enhancement of apoptosis (survivin, p53, caspase 3). Thus, in conclusion, STG as adjunctive therapy for Dox could be a strategy for the treatment of breast cancer patients, by their ability in hindering cell proliferation and minimizing the associated oxidative and inflammatory adverse reactions.
Collapse
Affiliation(s)
- Mohamed M. Salama
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 35712, Egypt
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Randa A. Zaghloul
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Rania M. Khalil
- Department of Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa 35712, Egypt
| | | |
Collapse
|
11
|
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
|
12
|
Parmar HS, Nayak A, Kataria S, Tripathi V, Jaiswal P, Gavel PK, Jha HC, Bhagwat S, Dixit AK, Lukashevich V, Das AK, Sharma R. Restructuring the ONYX-015 adenovirus by using spike protein genes from SARS-CoV-2 and MERS-CoV: Possible implications in breast cancer treatment. Med Hypotheses 2022; 159:110750. [PMID: 35002022 PMCID: PMC8723760 DOI: 10.1016/j.mehy.2021.110750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 11/29/2022]
Affiliation(s)
| | - Aakruti Nayak
- School of Biotechnology, Devi Ahilya University, Indore 452001, M.P., India
| | - Shreya Kataria
- School of Biotechnology, Devi Ahilya University, Indore 452001, M.P., India
| | - Versha Tripathi
- School of Biotechnology, Devi Ahilya University, Indore 452001, M.P., India
| | - Pooja Jaiswal
- School of Biotechnology, Devi Ahilya University, Indore 452001, M.P., India
| | | | - Hem Chandra Jha
- Department of Bioscience and Bioengineering, IIT, Simrol, Indore, India
| | - Shivani Bhagwat
- Suraksha Diagnostics Pvt Ltd, Newtown, Rajarhat, Kolkata, West Bengal, India
| | - Amit Kumar Dixit
- Central Council for Research in Ayurvedic Sciences, Kolkata, West Bengal, India
| | - Vladimir Lukashevich
- Institute of Physiology of the National Academy of Sciences of Belarus, Minsk 220072, Belarus
| | - Apurba Kumar Das
- Department of Bioscience and Bioengineering, IIT, Simrol, Indore, India
| | - Rajesh Sharma
- School of Pharmacy, Devi Ahilya University, Indore 452001, M.P., India
| |
Collapse
|
13
|
Yuan W, Yong W, Zhu J, Shi D. DPP4 Regulates DHCR24-Mediated Cholesterol Biosynthesis to Promote Methotrexate Resistance in Gestational Trophoblastic Neoplastic Cells. Front Oncol 2021; 11:704024. [PMID: 34926239 PMCID: PMC8675944 DOI: 10.3389/fonc.2021.704024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 11/17/2021] [Indexed: 11/17/2022] Open
Abstract
Metabolic reprogramming could promote cellular adaptation in response to chemotherapeutic drugs in cancer cells. Herein, we aimed to characterize the metabolomic profiles regulated by Dipeptidyl Peptidase 4 (DPP4) in methotrexate (MTX)-resistant gestational trophoblastic neoplastic (GTN) cells. A total of eighty metabolites were found to be commonly altered in DPP4-depleted JAR/MTX and JEG3/MTX cells. Cholesterol biosynthesis-related metabolites were markedly impacted by DPP4 knockdown in MTX-resistant sublines. Manipulation of DPP4 expression remarkably affected the level of cellular cholesterol in GTN cells. Our analysis also identified 24-Dehydrocholesterol Reductase (DHCR24) as a potential downstream effector of DPP4. Manipulation of DHCR24 expression affected cellular cholesterol level, reactive oxygen species (ROS) accumulation, and chemosensitivity to MTX in GTN cell models. In addition, over-expression of DHCR24 could markedly restore cellular cholesterol level and rescue cell survival in DPP4-depleted MTX-resistant GTN cells. Highly correlated expression of DPP4 and DHCR24 was observed in clinical GTN specimens. Further, DPP4 inhibitor sitagliptin effectively inhibited cholesterol biosynthesis, reduced DHCR24 expression and enhanced MTX-induced cytotoxicity in vitro and in vivo. In conclusion, our findings suggested that DPP4 might regulate DHCR24-mediated cholesterol biosynthesis to promote methotrexate resistance in GTN cells. Targeting DPP4/DHCR24 signaling might help to sensitize MTX-resistant GTN to MTX treatment.
Collapse
Affiliation(s)
- Weijie Yuan
- Department of Gastrointestinal Surgery, Xiangya Hospital, Central South University, Changsha, China.,The Hunan Provincial Key Laboratory of Precision Diagnosis and Treatment for Gastrointestinal Tumor, Changsha, China
| | - Wenjing Yong
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Zhu
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, China
| | - Dazun Shi
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
14
|
Jaiswal P, Tripathi V, Nayak A, Kataria S, Lukashevich V, Das A, Parmar HS. A molecular link between diabetes and breast cancer: Therapeutic potential of repurposing incretin-based therapies for breast cancer. Curr Cancer Drug Targets 2021; 21:829-848. [PMID: 34468298 DOI: 10.2174/1568009621666210901101851] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 11/22/2022]
Abstract
Female breast cancer recently surpassed lung cancer and became the most commonly diagnosed cancer worldwide. As per the recent data from WHO, breast cancer accounts for one out of every 8 cancer cases diagnosed among an estimated 2.3 million new cancer cases. Breast cancer is the most prevailing cancer type among women causing the highest number of cancer-related mortality. It has been estimated that in 2020, 68,5000 women died due to this disease. Breast cancers have varying degrees of molecular heterogeneity; therefore, they are divided into various molecular clinical sub types. Recent reports suggest that type 2 diabetes (one of the common chronic diseases worldwide) is linked to the higher incidence, accelerated progression, and aggressiveness of different cancers; especially breast cancer. Breast cancer is hormone-dependent in nature and has a cross-talk with metabolism. A number of antidiabetic therapies are known to exert beneficial effects on various types of cancers, including breast cancer. However, only a few reports are available on the role of incretin-based antidiabetic therapies in cancer as a whole and in breast cancer in particular. The present review sheds light on the potential of incretin based therapies on breast cancer and explores the plausible underlying mechanisms. Additionally, we have also discussed the sub types of breast cancer as well as the intricate relationship between diabetes and breast cancer.
Collapse
Affiliation(s)
- Pooja Jaiswal
- School of Biotechnology, Devi Ahilya University, Indore-452001. M.P., India
| | - Versha Tripathi
- School of Biotechnology, Devi Ahilya University, Indore-452001. M.P., India
| | - Aakruti Nayak
- School of Biotechnology, Devi Ahilya University, Indore-452001. M.P., India
| | - Shreya Kataria
- School of Biotechnology, Devi Ahilya University, Indore-452001. M.P., India
| | - Vladimir Lukashevich
- Institute of Physiology of the National Academy of Sciences of Belarus, Minsk-220072. Belarus
| | - Apurba Das
- Department of Chemical Sciences, IIT, Indore, Simrol, Indore, M.P., India
| | | |
Collapse
|
15
|
Zhang J, Wu N, Shi D. The Involvement of the Mammalian Target of Rapamycin, Protein Tyrosine Phosphatase 1b and Dipeptidase 4 Signaling Pathways in Cancer and Diabetes: A Narrative Review. Mini Rev Med Chem 2021; 21:803-815. [PMID: 33185160 DOI: 10.2174/1389557520666201113110406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 05/30/2020] [Accepted: 07/20/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND The mammalian target of rapamycin (mTOR), protein tyrosine phosphatase 1b (PTP1B) and dipeptidase 4 (DPP4) signaling pathways regulate eukaryotic cell proliferation and metabolism. Previous researches described different transduction mechanisms in the progression of cancer and diabetes. METHODOLOGY We reviewed recent advances in the signal transduction pathways of mTOR, PTP1B and DPP4 regulation and determined the crosstalk and common pathway in diabetes and cancer. RESULTS We showed that according to numerous past studies, the proteins participate in the signaling networks for both diseases. CONCLUSION There are common pathways and specific proteins involved in diabetes and cancer. This article demonstrates and explains the potential mechanisms of association and future prospects for targeting these proteins in pharmacological studies.
Collapse
Affiliation(s)
- Jiajia Zhang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, Shandong, China
| | - Ning Wu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, Shandong, China
| | - Dayong Shi
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, Shandong, China
| |
Collapse
|
16
|
Cai Z, Lu C, He J, Liu L, Zou Y, Zhang Z, Zhu Z, Ge X, Wu A, Jiang T, Zheng H, Peng Y. Identification and characterization of circRNAs encoded by MERS-CoV, SARS-CoV-1 and SARS-CoV-2. Brief Bioinform 2021; 22:1297-1308. [PMID: 33757279 PMCID: PMC7799257 DOI: 10.1093/bib/bbaa334] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/28/2020] [Accepted: 10/23/2020] [Indexed: 12/17/2022] Open
Abstract
The life-threatening coronaviruses MERS-CoV, SARS-CoV-1 and SARS-CoV-2 (SARS-CoV-1/2) have caused and will continue to cause enormous morbidity and mortality to humans. Virus-encoded noncoding RNAs are poorly understood in coronaviruses. Data mining of viral-infection-related RNA-sequencing data has resulted in the identification of 28 754, 720 and 3437 circRNAs encoded by MERS-CoV, SARS-CoV-1 and SARS-CoV-2, respectively. MERS-CoV exhibits much more prominent ability to encode circRNAs in all genomic regions than those of SARS-CoV-1/2. Viral circRNAs typically exhibit low expression levels. Moreover, majority of the viral circRNAs exhibit expressions only in the late stage of viral infection. Analysis of the competitive interactions of viral circRNAs, human miRNAs and mRNAs in MERS-CoV infections reveals that viral circRNAs up-regulated genes related to mRNA splicing and processing in the early stage of viral infection, and regulated genes involved in diverse functions including cancer, metabolism, autophagy, viral infection in the late stage of viral infection. Similar analysis in SARS-CoV-2 infections reveals that its viral circRNAs down-regulated genes associated with metabolic processes of cholesterol, alcohol, fatty acid and up-regulated genes associated with cellular responses to oxidative stress in the late stage of viral infection. A few genes regulated by viral circRNAs from both MERS-CoV and SARS-CoV-2 were enriched in several biological processes such as response to reactive oxygen and centrosome localization. This study provides the first glimpse into viral circRNAs in three deadly coronaviruses and would serve as a valuable resource for further studies of circRNAs in coronaviruses.
Collapse
Affiliation(s)
- Zena Cai
- College of Biology, Hunan University
| | - Congyu Lu
- College of Biology, Hunan University
| | | | - Li Liu
- Hunan Yuelu mountain data science and Technology Research Institute Co., Ltd, Changsha, China
| | | | | | | | - Xingyi Ge
- College of Biology, Hunan University
| | - Aiping Wu
- Suzhou Institute of Systems Medicine
| | | | | | | |
Collapse
|
17
|
Subbarayan K, Ulagappan K, Wickenhauser C, Bachmann M, Seliger B. Immune Interaction Map of Human SARS-CoV-2 Target Genes: Implications for Therapeutic Avenues. Front Immunol 2021; 12:597399. [PMID: 33796097 PMCID: PMC8007772 DOI: 10.3389/fimmu.2021.597399] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 02/18/2021] [Indexed: 01/16/2023] Open
Abstract
There exists increasing evidence that people with preceding medical conditions, such as diabetes and cancer, have a higher risk of infection with SARS-CoV-2 and are more vulnerable to severe disease. To get insights into the possible role of the immune system upon COVID-19 infection, 2811 genes of the gene ontology term “immune system process GO: 0002376” were selected for coexpression analysis of the human targets of SARS-CoV-2 (HT-SARS-CoV-2) ACE2, TMPRSS2, and FURIN in tissue samples from patients with cancer and diabetes mellitus. The network between HT-SARS-CoV-2 and immune system process genes was analyzed based on functional protein associations using STRING. In addition, STITCH was employed to determine druggable targets. DPP4 was the only immune system process gene, which was coexpressed with the three HT-SARS-CoV-2 genes, while eight other immune genes were at least coexpressed with two HT-SARS-CoV-2 genes. STRING analysis between immune and HT-SARS-CoV-2 genes plotted 19 associations of which there were eight common networking genes in mixed healthy (323) and pan-cancer (11003) tissues in addition to normal (87), cancer (90), and diabetic (128) pancreatic tissues. Using this approach, three commonly applicable druggable connections between HT-SARS-CoV-2 and immune system process genes were identified. These include positive associations of ACE2—DPP4 and TMPRSS2—SRC as well as a negative association of FURIN with ADAM17. Furthermore, 16 drugs were extracted from STITCH (score <0.8) with 32 target genes. Thus, an immunological network associated with HT-SARS-CoV-2 using bioinformatics tools was identified leading to novel therapeutic opportunities for COVID-19.
Collapse
Affiliation(s)
- Karthikeyan Subbarayan
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Kamatchi Ulagappan
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Claudia Wickenhauser
- Institute of Pathology, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Michael Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,National Center for Tumor Diseases, University Hospital "Carl Gustav Carus," Technische Universität (TU) Dresden, Dresden, Germany.,Tumor Immunology, University Cancer Center "Carl Gustav Carus," Technische Universität (TU) Dresden, Dresden, Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle, Germany.,Cell therapy and Immunology, Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| |
Collapse
|
18
|
Wilson AL, Moffitt LR, Wilson KL, Bilandzic M, Wright MD, Gorrell MD, Oehler MK, Plebanski M, Stephens AN. DPP4 Inhibitor Sitagliptin Enhances Lymphocyte Recruitment and Prolongs Survival in a Syngeneic Ovarian Cancer Mouse Model. Cancers (Basel) 2021; 13:487. [PMID: 33513866 PMCID: PMC7865851 DOI: 10.3390/cancers13030487] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/12/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023] Open
Abstract
Immunity plays a key role in epithelial ovarian cancer (EOC) progression with a well-documented correlation between patient survival and high intratumoral CD8+ to T regulatory cell (Treg) ratios. We previously identified dysregulated DPP4 activity in EOCs as a potentially immune-disruptive influence contributing to a reduction in CXCR3-mediated T-cell infiltration in solid tumours. We therefore hypothesized that inhibition of DPP4 activity by sitagliptin, an FDA-approved inhibitor, would improve T-cell infiltration and function in a syngeneic ID8 mouse model of EOC. Daily oral sitagliptin at 50 mg/kg was provided to mice with established primary EOCs. Sitagliptin treatment decreased metastatic tumour burden and significantly increased overall survival and was associated with significant changes to the immune landscape. Sitagliptin increased overall CXCR3-mediated CD8+ T-cell trafficking to the tumour and enhanced the activation and proliferation of CD8+ T-cells in tumour tissue and the peritoneal cavity. Substantial reductions in suppressive cytokines, including CCL2, CCL17, CCL22 and IL-10, were also noted and were associated with reduced CD4+ CD25+ Foxp3+ Treg recruitment in the tumour. Combination therapy with paclitaxel, however, typical of standard-of-care for patients in palliative care, abolished CXCR3-specific T-cell recruitment stimulated by sitagliptin. Our data suggest that sitagliptin may be suitable as an adjunct therapy for patients between chemotherapy cycles as a novel approach to enhance immunity, optimise T-cell-mediated function and improve overall survival.
Collapse
Affiliation(s)
- Amy L. Wilson
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton 3168, Australia; (A.L.W.); (L.R.M.); (M.B.)
- Department of Molecular and Translational Sciences, Monash Health, Clayton 3168, Australia
- Department of Immunology and Pathology, Monash University, Clayton 3800, Australia;
| | - Laura R. Moffitt
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton 3168, Australia; (A.L.W.); (L.R.M.); (M.B.)
- Department of Molecular and Translational Sciences, Monash Health, Clayton 3168, Australia
| | - Kirsty L. Wilson
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia;
| | - Maree Bilandzic
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton 3168, Australia; (A.L.W.); (L.R.M.); (M.B.)
- Department of Molecular and Translational Sciences, Monash Health, Clayton 3168, Australia
| | - Mark D. Wright
- Department of Immunology and Pathology, Monash University, Clayton 3800, Australia;
| | - Mark D. Gorrell
- Centenary Institute, Faculty of Medicine and Health, University of Sydney, Camperdown 2006, Australia;
| | - Martin K. Oehler
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide 5000, Australia;
| | - Magdalena Plebanski
- School of Health and Biomedical Sciences, RMIT University, Bundoora 3083, Australia;
| | - Andrew N. Stephens
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton 3168, Australia; (A.L.W.); (L.R.M.); (M.B.)
- Department of Molecular and Translational Sciences, Monash Health, Clayton 3168, Australia
| |
Collapse
|
19
|
Gomaa SH, Abaza MM, Elattar HA, Amin GA, Elshahawy DM. Soluble cluster of differentiation 26/soluble dipeptidyl peptidase-4 and glypican-3 are promising serum biomarkers for the early detection of Hepatitis C virus related hepatocellular carcinoma in Egyptians. Arab J Gastroenterol 2020; 21:224-232. [PMID: 32891543 DOI: 10.1016/j.ajg.2020.04.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 02/05/2020] [Accepted: 04/19/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND STUDY AIMS Many patients are diagnosed with hepatocellular carcinoma (HCC) in the late stage when it is already untreatable. Therefore, there is an increased need for sensitive biomarkers to detect HCC at an earlier stage in high risk patients with hepatitis C virus (HCV)-induced cirrhosis. This study aimed to evaluate the diagnostic performance of soluble cluster of differentiation 26/dipeptidyl peptidase 4 (sCD26/sDPP4) and glypican-3 (GPC3) as serum biomarkers for the early detection of HCV related HCC and compare it with that of the conventional tumor marker serum alpha fetoprotein (AFP). PATIENTS AND METHODS The study included 80 participants, 30 patients diagnosed with HCV infection without HCC (HCV group), 30 patients diagnosed with HCV- related HCC (HCV group), and 20 healthy volunteers (control group). The serum levels of GPC3 and sCD26 were measured using specific enzyme linked immunosorbant assay (ELISA) kits, whereas AFP levels were determined using chemiluminescence. RESULTS The serum levels of both sCD26 and GPC3 were found to be significantly higher in patients with early-stage HCC than in the HCV group, (1450 and 1.16 ng/mL, respectively). sCD26 at a cutoff value of > 1000 ng/ml, showed a high sensitivity (83.3%) and 63.3% specificity with an area under curve (AUC) of 0.811 and a 95% confidence interval (CI) of (0.682-0.94). While, the combination of GPC3 and sCD26 exhibited the best diagnostic performance for early-stage-HCC because it increased the sensitivity and specificity (85% and 93.3% respectively), with an AUC of 0.986 and a 95% CI of (0.899-1.00) compared to sCD26 alone. CONCLUSION We conclude that serum sCD26 could be a sensitive biomarker for the early detection of HCC among HCV patients. Moreover, the combination of sCD26 and GPC3 increases both the sensitivity and specificity for the early detection of HCV related HCC compared with AFP and could help in the monitoring of HCC in high risk patients with HCV induced cirrhosis.
Collapse
Affiliation(s)
- Salwa H Gomaa
- Department of Chemical Pathology, Medical Research Institute, Alexandria University, Egypt.
| | - Mona M Abaza
- Department of Chemical Pathology, Medical Research Institute, Alexandria University, Egypt
| | - Hoda A Elattar
- Department of Chemical Pathology, Medical Research Institute, Alexandria University, Egypt
| | - Gamal A Amin
- Department of Experimental and Internal Medicine, Medical Research Institute, Alexandria University, Egypt
| | | |
Collapse
|
20
|
Qiu L, Ma Y, Yang Y, Ren X, Wang D, Jia X. Pro-Angiogenic and Pro-Inflammatory Regulation by lncRNA MCM3AP-AS1-Mediated Upregulation of DPP4 in Clear Cell Renal Cell Carcinoma. Front Oncol 2020; 10:705. [PMID: 32714856 PMCID: PMC7344272 DOI: 10.3389/fonc.2020.00705] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/15/2020] [Indexed: 12/20/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) represents the most common type of renal cell carcinoma (RCC) in adults, in addition to the worst prognosis among the common epithelial kidney tumors. Inflammation and angiogenesis seem to potentiate tumor growth and metastasis of the malignancy. The current study explored the contributions of the lncRNA MCM3AP-AS1 in tumor-associated inflammation and angiogenesis in ccRCC with a specific focus on its transcriptional regulation and its interactions with transcription factor E2F1 and DPP4. Tumor tissues and matched adjacent non-tumor tissues were collected from 78 ccRCC patients. Methylation-specific PCR and ChIP assays were applied to detect the methylation at the promoter region of MCM3AP-AS1. Dual-luciferase reporter assay, RIP, RNA pull-down, and ChIP assays were employed to confirm the interactions between MCM3AP-AS1, E2F1, and DPP4. Nude mice were subcutaneously xenografted with human ccRCC cells. Cell proliferation was evaluated by CCK-8 assays and EDU staining in ccRCC cells in vitro and by immunohistochemical staining of Ki67 in vivo. Inflammation was examined by detecting the secretion of pro-inflammatory cytokines (TNF-α, IL-1β, and IL-6). Pro-angiogenic ability of ccRCC cells was assessed by the co-culture with human umbilical vein endothelial cells (HUVEC) in vitro and by microvessel density (MVD) measurements and angiogenesis in the chicken chorioallantoic membrane. MCM3AP-AS1 was highly-expressed in ccRCC and associated with poor patient survival. Demethylation of MCM3AP-AS1 was noted in ccRCC tissues and cells. Over-expression of MCM3AP-AS1 enhanced cell proliferation, the release of pro-inflammatory cytokines, and the tube formation of HUVECs in cultured human Caki-1 and 786-O cells. MCM3AP-AS1 was shown to enhance the E2F1 enrichment at the DPP4 promoter, to further increase the expression of DPP4. Knockdown of DPP4 could abate pro-angiogenic and pro-inflammatory abilities of MCM3AP-AS1 in ccRCC cells. Pro-angiogenic and pro-inflammatory abilities of MCM3AP-AS1 in vivo were confirmed in mice subcutaneously xenografted with human ccRCC cells. Our findings demonstrate a novel mechanism by which lncRNA MCM3AP-AS1 exerts pro-angiogenic and pro-inflammatory effects, highlighting the potential of MCM3AP-AS1 as a promising target for treating ccRCC.
Collapse
Affiliation(s)
- Ling Qiu
- Department of Tumor Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Yan Ma
- Department of Tumor Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Yanming Yang
- Department of Tumor Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Xiaojun Ren
- Department of Tumor Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Dongzhou Wang
- Department of Tumor Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Xiaojing Jia
- Department of Tumor Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
21
|
Kirtonia A, Gala K, Fernandes SG, Pandya G, Pandey AK, Sethi G, Khattar E, Garg M. Repurposing of drugs: An attractive pharmacological strategy for cancer therapeutics. Semin Cancer Biol 2020; 68:258-278. [PMID: 32380233 DOI: 10.1016/j.semcancer.2020.04.006] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/20/2020] [Accepted: 04/22/2020] [Indexed: 02/07/2023]
Abstract
Human malignancies are one of the major health-related issues though out the world and anticipated to rise in the future. The development of novel drugs/agents requires a huge amount of cost and time that represents a major challenge for drug discovery. In the last three decades, the number of FDA approved drugs has dropped down and this led to increasing interest in drug reposition or repurposing. The present review focuses on recent concepts and therapeutic opportunities for the utilization of antidiabetics, antibiotics, antifungal, anti-inflammatory, antipsychotic, PDE inhibitors and estrogen receptor antagonist, Antabuse, antiparasitic and cardiovascular agents/drugs as an alternative approach against human malignancies. The repurposing of approved non-cancerous drugs is an effective strategy to develop new therapeutic options for the treatment of cancer patients at an affordable cost in clinics. In the current scenario, most of the countries throughout the globe are unable to meet the medical needs of cancer patients because of the high cost of the available cancerous drugs. Some of these drugs displayed potential anti-cancer activity in preclinic and clinical studies by regulating several key molecular mechanisms and oncogenic pathways in human malignancies. The emerging pieces of evidence indicate that repurposing of drugs is crucial to the faster and cheaper discovery of anti-cancerous drugs.
Collapse
Affiliation(s)
- Anuradha Kirtonia
- Amity Institute of Molecular Medicine and Stem cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida, 201313, India; Equal contribution
| | - Kavita Gala
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be University), Vile Parle West, Mumbai, 400056, India; Equal contribution
| | - Stina George Fernandes
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be University), Vile Parle West, Mumbai, 400056, India; Equal contribution
| | - Gouri Pandya
- Amity Institute of Molecular Medicine and Stem cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida, 201313, India; Equal contribution
| | - Amit Kumar Pandey
- Amity Institute of Biotechnology, Amity University Haryana, Manesar, Haryana, 122413, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Ekta Khattar
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be University), Vile Parle West, Mumbai, 400056, India.
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida, 201313, India.
| |
Collapse
|
22
|
Shah C, Hong YR, Bishnoi R, Ali A, Skelton WP, Dang LH, Huo J, Dang NH. Impact of DPP4 Inhibitors in Survival of Patients With Prostate, Pancreas, and Breast Cancer. Front Oncol 2020; 10:405. [PMID: 32296640 PMCID: PMC7136489 DOI: 10.3389/fonc.2020.00405] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 03/09/2020] [Indexed: 12/02/2022] Open
Abstract
Background: Dipeptidyl peptidase-4 (DPP4), a cell surface protein, exhibits a crucial role in tumor biology and regulation of the immune system. We aim to study the impact of DPP4 inhibitors (DPP4i) in patients with prostate cancer (PRC), pancreatic cancer (PC) and breast cancer (BC). Methods: Using the SEER and Medicare linked database, we identified patients with PRC or PC or BC with coexisting type II diabetes mellitus between 2007 and 2015. Patients were classified into four groups: (1) not on either DPP4i or metformin (reference group), this group included patient that were on anti-diabetic agents other than metformin or DPP4i (2) metformin only, (3) DPP4i only, and (4) DPP4i along with metformin (combination group). Overall survival (OS) analyses were performed using SAS®, version 9.4. Results: We identified 15,330 patients with PRC, 5,359 patients with PC and 16,085 patients with BC. In PRC cohort, patients on DPP4i had significant survival advantage with HR 0.77 (95% CI: 0.64–0.93), P = 0.005 when compared to the reference group. Patients taking metformin also had significant OS benefit with HR 0.87 (95% CI: 0.81–0.93), P < 0.0001 when compared to the reference group. However, in BC cohort, OS did not favor the patients taking DPP4i with HR 1.07 (95% CI: 0.93–1.25, P = 0.33). Similarly, in PC cohort, OS was indifferent for the patients on DPP4i with HR 1.07 (95% CI: 0.93–1.24, P = 0.68). Upon subgroup analyses of PRC patients, the survival favored the group taking DPP4i, irrespective of stage, use of chemotherapy, androgen-deprivation therapy, and prostatectomy or radiation therapy. Conclusions: DPP4i seems to improve survival in PRC patients; however, not in PC or BC patients. While the exact mechanism involved remains to be elucidated, a prospective clinical trial would help to confirm these findings.
Collapse
Affiliation(s)
- Chintan Shah
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL, United States
| | - Young-Rock Hong
- Department of Health Services Research, Management and Policy, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Rohit Bishnoi
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL, United States
| | - Azka Ali
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL, United States
| | - William Paul Skelton
- Division of Medical Oncology, H. Lee Moffitt Cancer Center, University of South Florida, Tampa, FL, United States
| | - Long H Dang
- Ochsner Medical Center, Baton Rouge, LA, United States
| | - Jinhai Huo
- Department of Health Services Research, Management and Policy, College of Public Health and Health Professions, University of Florida, Gainesville, FL, United States
| | - Nam H Dang
- Division of Hematology and Oncology, Department of Medicine, University of Florida, Gainesville, FL, United States
| |
Collapse
|
23
|
Kawaguchi T, Nakano D, Koga H, Torimura T. Effects of a DPP4 Inhibitor on Progression of NASH-related HCC and the p62/ Keap1/Nrf2-Pentose Phosphate Pathway in a Mouse Model. Liver Cancer 2019; 8:359-372. [PMID: 31768345 PMCID: PMC6873068 DOI: 10.1159/000491763] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 06/29/2018] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND AND AIMS Diabetes mellitus is a risk factor for hepatocellular carcinoma (HCC) in patients with nonalcoholic steatohepatitis (NASH). Dipeptidyl peptidase-4 inhibitor (DPP4i), an antidiabetic agent, is reported to affect cell proliferation. We aimed to investigate the effects of DPP4i on the progression of NASH-related HCC and its metabolic pathway in a mouse model. METHODS A mouse model of NASH-related HCC was used in this study. Eight-week-old mice were administered either DPP4i (sitagliptin 30 mg/kg/day; DPP4i group; n = 8) or distilled water (control group; n = 8) for 10 weeks. Then, HCC progression was evaluated by computed tomography. Changes in metabolites of HCC tissue were analyzed by metabolomic analysis. The localization and expression of p62, Keap1, Nrf2, and MCM7 were evaluated by immunostaining and immunoblotting, respectively. RESULTS The number and volume of HCC were significantly lower in the DPP4i group than in the control group (1.8 ± 1.2 vs. 4.5 ± 1.7/liver, p < 0.01; 11.2 ± 20.8 vs. 37.5 ± 72.5 mm3/tumor, p < 0.05). Metabolome analysis revealed that DPP4i significantly increased 6-phosphogluconic acid and ribose 5-phosphate levels and decreased the AMP-to-adenine and GMP-to-guanine ratios (AMP-to-adenine ratio 0.7 ± 0.2 vs. 2.0 ± 1.2, p < 0.01; GMP-to-guanine ratio 0.6 ± 0.3 vs. 1.5 ± 0.7, p < 0.01). Immunostaining showed that p62 was localized in the cytoplasm of HCC in the DPP4i group, while p62 was localized in the nucleus of HCC in the control group. Keap1, Nrf2, and MCM7 expression decreased significantly in the DPP4i group compared to that in the control group. CONCLUSIONS We demonstrated that DDP4i prevented the progression of NASH-related HCC in a mouse model. Furthermore, metabolome analysis revealed that DDP4i downregulated the pentose phosphate pathway with suppression of the p62/Keap1/Nrf2 pathway. Thus, DDP4i may prevent tumor progression through inhibition of metabolic reprogramming in NASH-related HCC.
Collapse
Affiliation(s)
- Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan,*Takumi Kawaguchi, MD, PhD, Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume 830-0011 (Japan), E-Mail
| | - Dan Nakano
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Hironori Koga
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan,Liver Cancer Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| | - Takuji Torimura
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan,Liver Cancer Division, Research Center for Innovative Cancer Therapy, Kurume University, Kurume, Japan
| |
Collapse
|
24
|
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: 105] [Impact Index Per Article: 17.5] [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
|
25
|
Zhang N, Cong X, Zhou D, Guo L, Yuan C, Xu D, Su C. Predictive significance of serum dipeptidyl peptidase-IV in papillary thyroid carcinoma. Cancer Biomark 2019; 24:7-17. [PMID: 30594915 DOI: 10.3233/cbm-170908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND The significance of serum dipeptidyl peptidase-IV (DPP-IV) in papillary thyroid carcinoma (PTC) has not been elucidated. OBJECTIVE This study aimed to assess the role of serum DPP-IV in the carcinogenesis and prognosis of PTC. METHODS The serum DPP-IV concentration was measured in 171 male patients with PTC, 81 male patients with a benign thyroid nodule (BTN), and 52 male healthy controls (HCs). Multivariate logistic regression and Cox regression analyses were performed to evaluate the correlations between variables. Receiver operating characteristic (ROC) curves were used to calculate the diagnosis accuracy. RESULTS The ROC curve indicated a good performance of DPP-IV for discriminating PTC from BTN, with an area under the curve (AUC) of 0.881 (95% CI, 0.840-0.922). Serum DPP-IV demonstrated a modest performance in predicting nonstructurally persistent disease/recurrent disease (NSPRD) survival, with an AUC of 0.778 (95% CI, 0.635-0.922). A serum DPP-IV level ⩾ 250 nkat/L (HR, 6.529; 95% CI, 2.090-20.398; P= 0.001) and an advanced tumor, lymph node, metastasis (TNM) stage (HR, 4.677; 95% CI, 1.498-14.605; P= 0.008) were found to be independent factors for predicting SPRD. PTC patients with a DPP-IV level ⩾ 250 nkat/L had a worse outcome than those with a DPP-IV level < 250 nkat/L (P< 0.001). CONCLUSIONS Serum DPP-IV may be a predictive biomarker for PTC diagnosis and prognosis in Chinese male patients.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Xiaoqiang Cong
- Department of Cardiology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Dan Zhou
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Liang Guo
- Department of Pathology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Congwang Yuan
- Department of Pain, Yancheng First People's Hospital, Yancheng, Jiangsu 224000, China
| | - Dahai Xu
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin 130021, China
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Chang Su
- Department of Thyroid Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
26
|
Celastrol strongly inhibits proliferation, migration and cancer stem cell properties through suppression of Pin1 in ovarian cancer cells. Eur J Pharmacol 2019; 842:146-156. [DOI: 10.1016/j.ejphar.2018.10.043] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 10/22/2018] [Accepted: 10/29/2018] [Indexed: 12/25/2022]
|
27
|
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
|
28
|
Ma JQ, Yang Y, Juan J, Guo CF, Tuerxun M, Ting W, Hasim A. Over-expression of prolyl isomerase Pin1 promotes cervical tumorigenesis and metastasis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:664-674. [PMID: 31938152 PMCID: PMC6958022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 10/30/2017] [Indexed: 06/10/2023]
Abstract
Overexpression of the prolyl isomerase PIN1 is involved in tumorigenesis, but the role of PIN1 in cervical cancer is unclear. In this study, we examined PIN1 protein expression by immunohistochemistry in 221 paraffin-embedded samples from cervical cancer patients, cervical intraepithelial neoplasia patients, and control tissues, and found that high expression of PIN1 was significantly associated with lymph node metastasis (P=0.002), advanced stage according to the International Federation of Gynecology and Obstetrics guidelines (P=0.026). When endogenous PIN1 expression was knocked down using siRNA, cell proliferation, colony formation, migration, and invasion were inhibited in the SiHa cervical cancer cell line. Additionally, PIN1 knockdown increased E-cadherin and β-catenin expression, and decreased expression of N-cadherin and Vimentin, suggesting that PIN1 can promote epithelial-mesenchymal transition (EMT). These results indicate that the Overexpression of the prolyl isomerase PIN1 in cervical cancer indicates tumor-Promotive properties of PIN1 that may be a marker of poor prognosis in cervical cancer patients, and the molecular determinants of epithelial polarity which have tumorigenesis enhancing impact, might through EMT.
Collapse
Affiliation(s)
- Jun-Qi Ma
- Department of Gynecology, The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, China
| | - Yun Yang
- Department of Pathology, Medical University of XinjiangUrumqi, China
| | - Jiao Juan
- Department of Pathology, Medical University of XinjiangUrumqi, China
| | - Chun-Feng Guo
- Department of Gynecology, The First Affiliated Hospital of Xinjiang Medical UniversityUrumqi, China
| | - Meiliban Tuerxun
- Department of Pathology, Medical University of XinjiangUrumqi, China
| | - Wang Ting
- Library of Xinjiang Medical UniversityXinjiang, China
| | - Ayshamgul Hasim
- Department of Pathology, Medical University of XinjiangUrumqi, China
| |
Collapse
|
29
|
Qin CJ, Zhao LH, Zhou X, Zhang HL, Wen W, Tang L, Zeng M, Wang MD, Fu GB, Huang S, Huang WJ, Yang Y, Bao ZJ, Zhou WP, Wang HY, Yan HX. Inhibition of dipeptidyl peptidase IV prevents high fat diet-induced liver cancer angiogenesis by downregulating chemokine ligand 2. Cancer Lett 2018; 420:26-37. [PMID: 29409972 DOI: 10.1016/j.canlet.2018.01.064] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 01/20/2018] [Accepted: 01/22/2018] [Indexed: 12/12/2022]
Abstract
Obesity is a major risk factor for hepatocellular carcinoma (HCC) and is typically accompanied by higher levels of serum dipeptidyl peptidase 4 (DPP4). However, the role of DPP4 in obesity-promoted HCC is unclear. Here, we found that consumption of a high-fat diet (HFD) promoted HCC cell proliferation and metastasis and led to poor survival in a carcinogen-induced model of HCC in rats. Notably, genetic ablation of DPP4 or treatment with a DPP4 inhibitor (vildagliptin) prevented HFD-induced HCC. Moreover, HFD-induced DPP4 activity facilitated angiogenesis and cancer cell metastasis in vitro and in vivo, and vildagliptin prevented tumor progression by mediating the pro-angiogenic role of chemokine ligand 2 (CCL2). Loss of DPP4 effectively reversed HFD-induced CCL2 production and angiogenesis, indicating that the DPP4/CCL2/angiogenesis cascade had key roles in HFD-associated HCC progression. Furthermore, concomitant changes in serum DPP4 and CCL2 were observed in 210 patients with HCC, and high serum DPP4 activity was associated with poor clinical prognosis. These results revealed a link between obesity-related high serum DPP4 activity and HCC progression. Inhibition of DPP4 may represent a novel therapeutic intervention for patients with HCC.
Collapse
Affiliation(s)
- Chen-Jie Qin
- International Cooperation Laboratory on Signal Transduction, Easten Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer Research, Shanghai 201805, China; Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Ling-Hao Zhao
- National Center for Liver Cancer Research, Shanghai 201805, China; The Third Department of Easten Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Xu Zhou
- International Cooperation Laboratory on Signal Transduction, Easten Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer Research, Shanghai 201805, China
| | - Hui-Lu Zhang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wen Wen
- International Cooperation Laboratory on Signal Transduction, Easten Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer Research, Shanghai 201805, China
| | - Liang Tang
- International Cooperation Laboratory on Signal Transduction, Easten Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer Research, Shanghai 201805, China
| | - Min Zeng
- International Cooperation Laboratory on Signal Transduction, Easten Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer Research, Shanghai 201805, China
| | - Ming-Da Wang
- International Cooperation Laboratory on Signal Transduction, Easten Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer Research, Shanghai 201805, China
| | - Gong-Bo Fu
- International Cooperation Laboratory on Signal Transduction, Easten Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer Research, Shanghai 201805, China
| | - Shuai Huang
- Department of Tumor Minimally Invasive Surgery, Reiji Hospital, Shanghai Jiaotong University, Shanghai 200127, China
| | - Wei-Jian Huang
- International Cooperation Laboratory on Signal Transduction, Easten Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer Research, Shanghai 201805, China
| | - Yuan Yang
- The Third Department of Easten Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Zhi-Jun Bao
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai 200040, China
| | - Wei-Ping Zhou
- National Center for Liver Cancer Research, Shanghai 201805, China; The Third Department of Easten Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China
| | - Hong-Yang Wang
- International Cooperation Laboratory on Signal Transduction, Easten Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer Research, Shanghai 201805, China.
| | - He-Xin Yan
- International Cooperation Laboratory on Signal Transduction, Easten Hepatobiliary Surgery Institute, The Second Military Medical University, Shanghai 200438, China; National Center for Liver Cancer Research, Shanghai 201805, China.
| |
Collapse
|
30
|
Dingenouts CKE, Bakker W, Lodder K, Wiesmeijer KC, Moerkamp AT, Maring JA, Arthur HM, Smits AM, Goumans MJ. Inhibiting DPP4 in a mouse model of HHT1 results in a shift towards regenerative macrophages and reduces fibrosis after myocardial infarction. PLoS One 2017; 12:e0189805. [PMID: 29253907 PMCID: PMC5734765 DOI: 10.1371/journal.pone.0189805] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 12/02/2017] [Indexed: 12/11/2022] Open
Abstract
AIMS Hereditary Hemorrhagic Telangiectasia type-1 (HHT1) is a genetic vascular disorder caused by haploinsufficiency of the TGFβ co-receptor endoglin. Dysfunctional homing of HHT1 mononuclear cells (MNCs) towards the infarcted myocardium hampers cardiac recovery. HHT1-MNCs have elevated expression of dipeptidyl peptidase-4 (DPP4/CD26), which inhibits recruitment of CXCR4-expressing MNCs by inactivation of stromal cell-derived factor 1 (SDF1). We hypothesize that inhibiting DPP4 will restore homing of HHT1-MNCs to the infarcted heart and improve cardiac recovery. METHODS AND RESULTS After inducing myocardial infarction (MI), wild type (WT) and endoglin heterozygous (Eng+/-) mice were treated for 5 days with the DPP4 inhibitor Diprotin A (DipA). DipA increased the number of CXCR4+ MNCs residing in the infarcted Eng+/- hearts (Eng+/- 73.17±12.67 vs. Eng+/- treated 157.00±11.61, P = 0.0003) and significantly reduced infarct size (Eng+/- 46.60±9.33% vs. Eng+/- treated 27.02±3.04%, P = 0.03). Echocardiography demonstrated that DipA treatment slightly deteriorated heart function in Eng+/- mice. An increased number of capillaries (Eng+/- 61.63±1.43 vs. Eng+/- treated 74.30±1.74, P = 0.001) were detected in the infarct border zone whereas the number of arteries was reduced (Eng+/- 11.88±0.63 vs. Eng+/- treated 6.38±0.97, P = 0.003). Interestingly, while less M2 regenerative macrophages were present in Eng+/- hearts prior to DipA treatment, (WT 29.88±1.52% vs. Eng+/- 12.34±1.64%, P<0.0001), DPP4 inhibition restored the number of M2 macrophages to wild type levels. CONCLUSIONS In this study, we demonstrate that systemic DPP4 inhibition restores the impaired MNC homing in Eng+/- animals post-MI, and enhances cardiac repair, which might be explained by restoring the balance between the inflammatory and regenerative macrophages present in the heart.
Collapse
Affiliation(s)
| | - Wineke Bakker
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Kirsten Lodder
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Karien C. Wiesmeijer
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Asja T. Moerkamp
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Janita A. Maring
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Helen M. Arthur
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, Newcastle upon Tyne, United Kingdom
| | - Anke M. Smits
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marie-José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
31
|
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: 42] [Impact Index Per Article: 5.3] [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
|
32
|
Lee JJ, Wang TY, Liu CL, Chien MN, Chen MJ, Hsu YC, Leung CH, Cheng SP. Dipeptidyl Peptidase IV as a Prognostic Marker and Therapeutic Target in Papillary Thyroid Carcinoma. J Clin Endocrinol Metab 2017; 102:2930-2940. [PMID: 28575350 DOI: 10.1210/jc.2017-00346] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 05/23/2017] [Indexed: 12/18/2022]
Abstract
CONTEXT Dipeptidyl peptidase IV (DPP4) is overexpressed in thyroid cancer and certain malignancies. Furthermore, DPP4 has been identified as a discriminatory marker for thyroid cancer. However, it remains unclear whether DPP4 expression plays a prognostic role. OBJECTIVE The aim of this study was to investigate the expression and function of DPP4 in thyroid cancer and the mechanisms involved. DESIGN We determined the expression of DPP4 by immunohistochemistry in tissue microarrays of thyroid tumors. In vitro functional studies were performed after genetic and pharmacological inhibition of DPP4. Gene expression and pathway analyses were used to identify downstream targets. The therapeutic potential of DPP4 inhibition was evaluated in a mouse xenograft model. RESULTS High DPP4 expression was associated with extrathyroidal extension (P < 0.001), BRAF mutation (P < 0.001), and advanced tumor stage (P = 0.007) in papillary thyroid cancer. Patients in the high-DPP4 expression group were less likely to be classified as having no evidence of disease at final follow-up (P = 0.042). DPP4 silencing or treatment with DPP4 inhibitors significantly suppressed colony formation, cell migration, and invasion. Analysis of differentially expressed genes after DPP4 knockdown suggested that the transforming growth factor-β signaling pathway is involved. In vivo experiments revealed that sitagliptin treatment reduced tumor growth and xenograft transforming growth factor-β receptor I expression. CONCLUSIONS Increased DPP4 expression is associated with cellular invasion and more aggressive disease in papillary thyroid cancer. Targeting DPP4 may be a therapeutic strategy for DPP4-expressing thyroid cancer.
Collapse
Affiliation(s)
- Jie-Jen Lee
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei 10449, Taiwan
- Graduate Institute of Medical Sciences and Department of Pharmacology, Taipei Medical University, Taipei 11031, Taiwan
| | - Tao-Yeuan Wang
- Department of Pathology, MacKay Memorial Hospital and Mackay Medical College, Taipei 10449, Taiwan
| | - Chien-Liang Liu
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei 10449, Taiwan
| | - Ming-Nan Chien
- Division of Endocrinology and Metabolism, Department of Internal Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei 10449, Taiwan
| | - Ming-Jen Chen
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei 10449, Taiwan
- Graduate Institute of Medical Sciences and Department of Pharmacology, Taipei Medical University, Taipei 11031, Taiwan
| | - Yi-Chiung Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City 32001, Taiwan
| | - Ching-Hsiang Leung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, MacKay Memorial Hospital and Mackay Medical College, Taipei 10449, Taiwan
| | - Shih-Ping Cheng
- Department of Surgery, MacKay Memorial Hospital and Mackay Medical College, Taipei 10449, Taiwan
- Graduate Institute of Medical Sciences and Department of Pharmacology, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
33
|
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
|
34
|
Wang XM, Holz LE, Chowdhury S, Cordoba SP, Evans KA, Gall MG, Vieira de Ribeiro AJ, Zheng YZ, Levy MT, Yu DM, Yao TW, Polak N, Jolly CJ, Bertolino P, McCaughan GW, Gorrell MD. The pro-fibrotic role of dipeptidyl peptidase 4 in carbon tetrachloride-induced experimental liver injury. Immunol Cell Biol 2017; 95:443-453. [PMID: 27899813 DOI: 10.1038/icb.2016.116] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 11/04/2016] [Accepted: 11/24/2016] [Indexed: 12/19/2022]
Abstract
Liver fibrosis is a progressive pathological process involving inflammation and extracellular matrix deposition. Dipeptidyl peptidase 4 (DPP4), also known as CD26, is a cell surface glycoprotein and serine protease. DPP4 binds to fibronectin, can inactivate specific chemokines, incretin hormone and neuropeptides, and influences cell adhesion and migration. Such properties suggest a pro-fibrotic role for this peptidase but this hypothesis needs in vivo examination. Experimental liver injury was induced with carbon tetrachloride (CCl4) in DPP4 gene knockout (gko) mice. DPP4 gko had less liver fibrosis and inflammation and fewer B cell clusters than wild type mice in the fibrosis model. DPP4 inhibitor-treated mice also developed less liver fibrosis. DNA microarray and PCR showed that many immunoglobulin (Ig) genes and some metabolism-associated transcripts were differentially expressed in the gko strain compared with wild type. CCl4-treated DPP4 gko livers had more IgM+ and IgG+ intrahepatic lymphocytes, and fewer CD4+, IgD+ and CD21+ intrahepatic lymphocytes. These data suggest that DPP4 is pro-fibrotic in CCl4-induced liver fibrosis and that the mechanisms of DPP4 pro-fibrotic action include energy metabolism, B cells, NK cells and CD4+ cells.
Collapse
Affiliation(s)
- Xin M Wang
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- A.W. Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Lauren E Holz
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Sumaiya Chowdhury
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Shaun P Cordoba
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Kathryn A Evans
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Margaret G Gall
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | | | - Yuan Zhou Zheng
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Miriam T Levy
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- A.W. Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Denise Mt Yu
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Tsun-Wen Yao
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Natasa Polak
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Christopher J Jolly
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Patrick Bertolino
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Geoffrey W McCaughan
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- A.W. Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Mark D Gorrell
- Centenary Institute and Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- A.W. Morrow Gastroenterology and Liver Centre, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| |
Collapse
|
35
|
Rathmann W, Kostev K. Association of dipeptidyl peptidase 4 inhibitors with risk of metastases in patients with type 2 diabetes and breast, prostate or digestive system cancer. J Diabetes Complications 2017; 31:687-692. [PMID: 28169131 DOI: 10.1016/j.jdiacomp.2017.01.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 01/20/2017] [Accepted: 01/22/2017] [Indexed: 12/27/2022]
Abstract
AIMS Experimental and animal studies have supported the hypothesis that dipeptidyl peptidase-4 inhibitors (DPP-4i) may accelerate tumor metastasis. The aim was to analyze the relationships between DPP-4i therapy with risk of metastases in type 2 diabetes patients with breast, prostate and digestive organ cancers. METHODS Type 2 diabetes patients with first diagnoses of breast, prostate or digestive organ cancer were selected in general and internal medicine practices (Disease Analyzer Germany: 01/2008-12/2014). Propensity score matching between DPP-4i users and non-users was carried out for age, sex, diabetes duration, and metformin use. Time-dependent Cox regression models were used to estimate hazard ratios (HR) for metastases further adjusting for HbA1c, body mass index, comorbidity and co-therapy with glucose-lowering drugs (3-4years follow-up). RESULTS 668 patients with newly diagnosed breast cancer, 906 with prostate cancer and 908 with digestive organ cancer were analyzed. In Cox regression, use of DPP-4i was not associated with an increased risk of metastases in patients with breast (adjusted HR, 95%CI: 1.00, 0.49-2.02), prostate (0.98, 0.54-1.77) or digestive organ cancers (0.97, 0.57-1.66). CONCLUSIONS This first observational study in patients with type 2 diabetes and breast, prostate or digestive organ cancer found no increased risk of metastases in DPP-4i users.
Collapse
Affiliation(s)
- Wolfgang Rathmann
- Institute of Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University, Düsseldorf, Germany
| | | |
Collapse
|
36
|
Henderson JM, Zhang HE, Polak N, Gorrell MD. Hepatocellular carcinoma: Mouse models and the potential roles of proteases. Cancer Lett 2017; 387:106-113. [PMID: 27045475 DOI: 10.1016/j.canlet.2016.03.047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 03/24/2016] [Accepted: 03/24/2016] [Indexed: 02/07/2023]
Abstract
Primary liver cancer is the second most common cause of mortality from cancer. The most common models of hepatocellular carcinoma, which use a chemical and/or metabolic insult, xenograft, or genetic manipulation, are discussed in this review. In the tumour microenvironment lymphocytes, fibroblasts, endothelial cells and antigen presenting cells are important determinants of cell fate. These cells make a range of proteases that modify the biological activity of other proteins, particularly extracellular matrix proteins that alter cell migration of tumour cells, fibroblasts and leucocytes, and chemokines that alter leucocyte migration. The DPP4 family of post-proline peptidase enzymes modifies cell movement and the activities of many bioactive molecules including growth factors and chemokines.
Collapse
Affiliation(s)
- James M Henderson
- Centenary Institute and Sydney Medical School, University of Sydney, Sydney, New South Wales 2006 Australia
| | - Hui Emma Zhang
- Centenary Institute and Sydney Medical School, University of Sydney, Sydney, New South Wales 2006 Australia
| | - Natasa Polak
- Centenary Institute and Sydney Medical School, University of Sydney, Sydney, New South Wales 2006 Australia
| | - Mark D Gorrell
- Centenary Institute and Sydney Medical School, University of Sydney, Sydney, New South Wales 2006 Australia.
| |
Collapse
|
37
|
Liang PI, Yeh BW, Li WM, Chan TC, Chang IW, Huang CN, Li CC, Ke HL, Yeh HC, Wu WJ, Li CF. DPP4/CD26 overexpression in urothelial carcinoma confers an independent prognostic impact and correlates with intrinsic biological aggressiveness. Oncotarget 2017; 8:2995-3008. [PMID: 27936466 PMCID: PMC5356858 DOI: 10.18632/oncotarget.13820] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 11/30/2016] [Indexed: 02/06/2023] Open
Abstract
Urothelial carcinoma (UC) is common cancer worldwide. The molecular aberrations regarding tumor progression remain unclear. Pericellular proteolysis is crucial in tumorigenesis, but its significance is unexplored in UC. By data mining the datasets in Gene Expression Omnibus, specifically focus on the proteolysis pathway, and followed by a preliminary validation in a pilot batch of tumor samples, we identified that the upregulation of dipeptidyl peptidase 4 (DPP4) was most significantly associated with clinical aggressiveness of UCs. Quantitative RT-PCR confirmed upregulation of DPP4 mRNA in advanced stage UCs. The clinical significance of DPP4 expression was validated in our large cohort consists of 635 UCs from upper urinary tract and urinary bladder. Univariate and multivariate analyses show that DPP4 is an independent prognosticatory biomarker for disease-specific survival and metastasis-free survival. Comparing the DPP4 expression level of three urothelial cell lines with normal urothelial cells, J82 and RTCC-1 showed a significantly increased in transcript and protein expression. DPP4 knockdown as conducted by using short-hairpin RNA resulted in a significantly decreased cell viability, proliferation, migration, and invasion in J82 and RTCC-1 cells. These findings implicate that DPP4 plays a role in the aggressiveness of UCs, and can serve as a novel prognostic marker and therapeutic target.
Collapse
Affiliation(s)
- Peir-In Liang
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Bi-Wen Yeh
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung, Medical University, Kaohsiung, Taiwan
| | - Wei-Ming Li
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung, Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Urology, Ministry of Health and Welfare Pingtung Hospital, Pingtung, Taiwan
| | - Ti-Chun Chan
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
- Institute of Biomedical Science, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - I-Wei Chang
- Department of Pathology, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
- School of Medicine for International Students, I-Shou University, Kaohsiung, Taiwan
| | - Chun-Nung Huang
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung, Medical University, Kaohsiung, Taiwan
| | - Ching-Chia Li
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung, Medical University, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Hung-Lung Ke
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung, Medical University, Kaohsiung, Taiwan
| | - Hsin-Chih Yeh
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung, Medical University, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Wen-Jeng Wu
- Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Kaohsiung, Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Urology, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
- Center for Infectious Disease and Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Stem Cell Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chien-Feng Li
- Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
- Department of Pathology, Chi Mei Medical Center, Tainan, Taiwan
- Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
- Institute of Clinical Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
38
|
Abstract
Targeted drugs have changed cancer treatment but are often ineffective in the long term against solid tumours, largely because of the activation of heterogeneous oncogenic pathways. A central common signalling mechanism in many of these pathways is proline-directed phosphorylation, which is regulated by many kinases and phosphatases. The structure and function of these phosphorylated proteins are further controlled by a single proline isomerase: PIN1. PIN1 is overactivated in cancers and it promotes cancer and cancer stem cells by disrupting the balance of oncogenes and tumour suppressors. This Review discusses the roles of PIN1 in cancer and the potential of PIN1 inhibitors to restore this balance.
Collapse
Affiliation(s)
- Xiao Zhen Zhou
- Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| | - Kun Ping Lu
- Division of Translational Therapeutics, Department of Medicine and Cancer Research Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, USA
| |
Collapse
|
39
|
Choi HJ, Kim JY, Lim SC, Kim G, Yun HJ, Choi HS. Dipeptidyl peptidase 4 promotes epithelial cell transformation and breast tumourigenesis via induction of PIN1 gene expression. Br J Pharmacol 2015; 172:5096-5109. [PMID: 26267432 PMCID: PMC4687806 DOI: 10.1111/bph.13274] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 06/19/2015] [Accepted: 07/10/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND PURPOSE Dipeptidyl peptidase 4 (DPP4) is an aminopeptidase that is widely expressed in different cell types. Recent studies suggested that DPP4 plays an important role in tumour progression in several human malignancies. Here we have examined the mechanisms by which up-regulation of DPP4 expression causes epithelial transformation and mammary tumourigenesis. EXPERIMENTAL APPROACH Expression of DPP4 and the peptidylprolyl cis/trans isomerase, NIMA-interacting 1 (PIN1), and the cytotoxic effects of combined treatment with sitagliptin and juglone were investigated by immunohistochemistry, immunoblotting, real-time PCR, TUNEL and soft agar assays, using MCF7 cells. The effects of sitagliptin on tumour development in vivo were studied in the syngeneic 4T1 metastatic breast cancer model. KEY RESULTS Activity of the transcription factor E2F1 induced by EGF was enhanced by DPP4, thus increasing PIN1 expression. Furthermore, DPP4 enhanced MEK/ERK and JNK/c-Jun signalling induced by EGF, inducing AP-1 activity and epithelial cell transformation. In contrast, DPP4 silencing or DPP4 inhibition in MCF7 cells inhibited PIN1 expression via E2F1 activity induced by EGF, decreasing colony formation and inducing DNA fragmentation. In the syngeneic 4T1 metastatic breast cancer model, DPP4 overexpression increased tumour development, whereas treatment with sitagliptin and/or juglone suppressed it. Consistent with these observations, DPP4 levels were positively correlated with PIN1 expression in human breast cancer. CONCLUSIONS AND IMPLICATIONS DPP4 promoted EGF-induced epithelial cell transformation and mammary tumourigenesis via induction of PIN1 expression, suggesting that sitagliptin targeting of DPP4 could be a treatment strategy in patients with breast cancer.
Collapse
Affiliation(s)
- H J Choi
- College of Pharmacy, Chosun University, Gwangju, South Korea
| | - J Y Kim
- College of Pharmacy, Chosun University, Gwangju, South Korea
| | - S-C Lim
- Department of Pathology, School of Medicine, Chosun University, Gwangju, South Korea
| | - G Kim
- College of Pharmacy, Chosun University, Gwangju, South Korea
| | - H J Yun
- College of Pharmacy, Chosun University, Gwangju, South Korea
| | - H S Choi
- College of Pharmacy, Chosun University, Gwangju, South Korea
| |
Collapse
|
40
|
Choi HJ, Kim JY, Lim SC, Kim G, Yun HJ, Choi HS. Dipeptidyl peptidase 4 promotes epithelial cell transformation and breast tumourigenesis via induction of PIN1 gene expression. Br J Pharmacol 2015. [PMID: 26267432 DOI: 10.1111/bph.13274.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Dipeptidyl peptidase 4 (DPP4) is an aminopeptidase that is widely expressed in different cell types. Recent studies suggested that DPP4 plays an important role in tumour progression in several human malignancies. Here we have examined the mechanisms by which up-regulation of DPP4 expression causes epithelial transformation and mammary tumourigenesis. EXPERIMENTAL APPROACH Expression of DPP4 and the peptidylprolyl cis/trans isomerase, NIMA-interacting 1 (PIN1), and the cytotoxic effects of combined treatment with sitagliptin and juglone were investigated by immunohistochemistry, immunoblotting, real-time PCR, TUNEL and soft agar assays, using MCF7 cells. The effects of sitagliptin on tumour development in vivo were studied in the syngeneic 4T1 metastatic breast cancer model. KEY RESULTS Activity of the transcription factor E2F1 induced by EGF was enhanced by DPP4, thus increasing PIN1 expression. Furthermore, DPP4 enhanced MEK/ERK and JNK/c-Jun signalling induced by EGF, inducing AP-1 activity and epithelial cell transformation. In contrast, DPP4 silencing or DPP4 inhibition in MCF7 cells inhibited PIN1 expression via E2F1 activity induced by EGF, decreasing colony formation and inducing DNA fragmentation. In the syngeneic 4T1 metastatic breast cancer model, DPP4 overexpression increased tumour development, whereas treatment with sitagliptin and/or juglone suppressed it. Consistent with these observations, DPP4 levels were positively correlated with PIN1 expression in human breast cancer. CONCLUSIONS AND IMPLICATIONS DPP4 promoted EGF-induced epithelial cell transformation and mammary tumourigenesis via induction of PIN1 expression, suggesting that sitagliptin targeting of DPP4 could be a treatment strategy in patients with breast cancer.
Collapse
Affiliation(s)
- H J Choi
- College of Pharmacy, Chosun University, Gwangju, South Korea
| | - J Y Kim
- College of Pharmacy, Chosun University, Gwangju, South Korea
| | - S-C Lim
- Department of Pathology, School of Medicine, Chosun University, Gwangju, South Korea
| | - G Kim
- College of Pharmacy, Chosun University, Gwangju, South Korea
| | - H J Yun
- College of Pharmacy, Chosun University, Gwangju, South Korea
| | - H S Choi
- College of Pharmacy, Chosun University, Gwangju, South Korea
| |
Collapse
|