1
|
Xie Q, Zhou J, He C, Xu Y, Tao F, Hu M. Unlocking the intricacies: Exploring the complex interplay between platelets and ovarian cancer. Crit Rev Oncol Hematol 2024; 202:104465. [PMID: 39097249 DOI: 10.1016/j.critrevonc.2024.104465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024] Open
Abstract
Ovarian cancer, an aggressive malignancy of the female reproductive tract, is frequently linked to an elevated risk of thrombotic events. This association is manifested by a pronounced rise in platelet counts and activation levels. Current research firmly supports the pivotal role of platelets in the oncogenic processes of ovarian cancer, influencing tumor cell proliferation and metastasis. Platelets influence these processes through direct interactions with tumor cells or by secreting cytokines and growth factors that enhance tumor growth, angiogenesis, and metastasis. This review aims to thoroughly dissect the interactions between platelets and ovarian cancer cells, emphasizing their combined role in tumor progression and associated thrombotic events. Additionally, it summarizes therapeutic strategies targeting platelet-cancer interface which show significant promise. Such approaches could not only be effective in managing the primary ovarian tumor but also play a pivotal role in preventing metastasis and attenuating thrombotic complications associated with ovarian cancer.
Collapse
Affiliation(s)
- Qianxin Xie
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie Zhou
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chaonan He
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ye Xu
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangfang Tao
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Mengjiao Hu
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
2
|
Karimi F, Dinarvand N, Sabaghan M, Azadbakht O, Ataee S, Kharazinejad E, Moazamfard M. Diabetes and ovarian cancer: risk factors, molecular mechanisms and impact on prognosis. Endocrine 2024; 83:1-9. [PMID: 37552417 DOI: 10.1007/s12020-023-03477-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND AND AIM Diabetes mellitus has been linked to a lower rate of cancer survival and an increase in the incidence of most malignancies. Investigations showed that diabetes might affect ovarian cancer (OC) prognosis and survival. Based on the current information, this study intends to review the risk factors, molecular pathways, and impact of diabetes on OC. METHODS The data was derived from online databases, including Web of Science, PubMed, and Scopus. The inclusion criteria were original studies, which included the risk factors, molecular mechanisms, and impact of diabetes on OC. The effect of different antidiabetic drugs was also discussed in this manuscript. All of the clinical, in vivo, and in vitro studies were included in the present study. RESULTS The diagnosis of diabetes mellitus negatively affects the survival and prognosis in OC cases. The epidemiologic data shows that the risk of OC increases in patients with diabetes mellitus compared to the healthy population. Insulin-like growth factors family was raised in diabetic patients, which target several mechanisms, including targeting oxidative stress, angiogenesis, and tumor markers. Antidiabetic drugs such as metformin, sitagliptin, and rosiglitazone have a promising effect on elongation of survival and enhancement of prognosis in OC patients. CONCLUSIONS Diabetes mellitus is a significant risk factor for OC in women, and it negatively impacts survival and prognosis. Molecular mechanisms such as IGF family, oxidative stress, and inflammatory cytokines have been identified to explain this relationship. Antidiabetic drugs like metformin, sitagliptin, and rosiglitazone have shown promise in improving survival and prognosis of OC patients.
Collapse
Affiliation(s)
- Farzaneh Karimi
- Department of Physiology, Behbahan Faculty of Medical Sciences, Behbahan, Iran.
| | - Negar Dinarvand
- Hyperlipidemia Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Sabaghan
- Department of Parasiotology, Behbahan Faculty of Medical Sciences, Behbahan, Iran
| | - Omid Azadbakht
- Department of Radiology Technology, Behbahan Faculty of Medical Sciences, Behbahan, Iran
| | - Shima Ataee
- Behbahan Faculty of Medical Sciences, Behbahan, Iran
| | - Ebrahim Kharazinejad
- Department of Anatomy, Faculty of Medicine, Abadan University of Medical Sciences, Abadan, Iran
| | - Mostafa Moazamfard
- Instructor of Operating Room, Behbahan Faculty of Medical Sciences, Behbahan, Iran.
| |
Collapse
|
3
|
Alvidrez RIM, Annarapu GK, Srinivasan AJ, Liu Z, Yazdani HO, Nolfi-Donegan D, Simmons RL, Shiva S, Neal MD. High Dose of Metformin Decreases Susceptibility to Occlusive Arterial Thrombosis in Diabetic Mice. JOURNAL OF PHARMACY AND PHARMACOLOGY RESEARCH 2023; 7:192-202. [PMID: 37975061 PMCID: PMC10653203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Introduction Metformin is the most prescribed medication in Diabetes Mellitus(DM). Metformin has shown to decrease mean platelet volume, with promising antiplatelet effects. High doses of Metformin have also been associated with hypercoagulation. We hypothesize that Metformin will protect DM mice from occlusive arterial thrombus formation by altering platelet activation and mitochondrial bioenergetics. Methods DM was developed by low dose of Streptozotocin, non-DM (healthy) mice are controls. Either vehicle or Metformin was administered twice daily via oral gavage for 7-days. Ferric chloride (FeCl3) arterial thrombosis and tail bleeding time were performed. Whole blood aggregometry, platelet activation/adhesion and mitochondrial bioenergetics were evaluated. Results Metformin decreased susceptibility of DM mice to arterial thrombosis. Platelet bioenergetics show DM mice have increased platelet mitochondrial respiration, but no differences were observed with Metformin treatment. In non-DM (healthy) mice, Metformin modulated ADP-dependent increase in platelet adhesion. Non-DM (healthy) mice, Metformin shortens bleeding time with faster thrombotic occlusion. Metformin also increased platelet mitochondrial maximal respiration and spare respiratory capacity uniquely in non-DM (healthy) mice. Conclusion Metformin regulates platelet bioenergetics and ADP-mediated platelet function in DM mice which attenuates susceptibility to arterial thrombosis. Future studies will evaluate clinically relevant doses of Metformin that regulates thrombotic function in diabetic platelets.
Collapse
Affiliation(s)
- Roberto I Mota Alvidrez
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA 15213
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
- Clinical and Translational Science Center, University of New Mexico, USA 87131
- Department of Pharmaceutical Sciences, University of New Mexico, USA 87131
| | - Gowtham K Annarapu
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
| | - Amudan J Srinivasan
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA 15213
| | - Zeyu Liu
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA 15213
- Department of Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
| | - Hamza O Yazdani
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA 15213
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
| | - Deidre Nolfi-Donegan
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
| | - Richard L Simmons
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA 15213
| | - Sruti Shiva
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
| | - Matthew D Neal
- Trauma and Transfusion Medicine Research Center, Department of Surgery, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA 15213
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, USA 15213
| |
Collapse
|
4
|
Lu MZ, Li DY, Wang XF. Effect of metformin use on the risk and prognosis of ovarian cancer: an updated systematic review and meta-analysis. Panminerva Med 2023; 65:351-361. [PMID: 31290300 DOI: 10.23736/s0031-0808.19.03640-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Emerging evidence suggests that metformin has a potential antitumor effect both in vitro and in vivo. Increasing epidemiological studies indicate that diabetic patients receiving metformin therapy have lower incidences of cancer and have better survival rates. However, there are limited and inconsistent studies available about the effect of metformin therapy on ovarian cancer (OC). Thus, we conducted this meta-analysis to study the effect of metformin therapy on OC. Meanwhile, we systematically reviewed relevant studies to provide a framework for future research. EVIDENCE ACQUISITION We conducted a systematic literature search on PubMed, Web of Science, Springerlink, CNKI, VIP, SinoMed, and Wanfang up to the period of October 2018. A random-effects meta-analysis model was used to derive pooled effect estimates. EVIDENCE SYNTHESIS A total of 13 studies were retrieved of which 5 studies explained the prevention and 8 studies explained the treatment for OC. Our pooled results showed that metformin has a potential preventive effect on OC in diabetic women (pooled odds ratio [OR] 0.62, 95% confidence interval [95% CI] 0.34, 1.11; P<0.001). In addition, metformin can also significantly prolong progression-free survival (PFS) (pooled hazard ratio [HR] 0.49, 95% CI 0.34, 0.70; P=0.002), and overall survival (OS) (HR 0.71, 95%CI 0.61, 0.82; P<0.001) in patients with OC, regardless of whether they had diabetes. CONCLUSIONS The use of metformin can potentially reduce the risk of OC among diabetics, and it also can significantly improve PFS and OS in patients with OC. A further large clinical investigation would be needed to adopt our finding in practice, however, our systematic review provides an insight for future study designs.
Collapse
Affiliation(s)
- Min-Zhen Lu
- Second Clinical Medical College of Southern Medical University, Guangzhou, China -
| | - De-Yu Li
- Department of Oncology, Fujian Provincial Hospital, Fujian, China
| | - Xue-Feng Wang
- Department of Obstetrics and Gynecology, Third Hospital of Southern Medical University, Guangzhou, China
| |
Collapse
|
5
|
Mota Alvidrez RI, Annarapu GK, Srinivasan AJ, Liu Z, Yazdani HO, Simmons RL, Shiva S, Neal MD, Nolfi-Donegan D. High Dose of Metformin Decreases Susceptibility to Occlusive Arterial Thrombosis in Diabetic Mice. RESEARCH SQUARE 2023:rs.3.rs-3143156. [PMID: 37503167 PMCID: PMC10371086 DOI: 10.21203/rs.3.rs-3143156/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Introduction Metformin is the most prescribed medication in Diabetes Mellitus(DM). Metformin has shown to decrease mean platelet volume, with promising antiplatelet effects. High doses of Metformin have also been associated with hypercoagulation. We hypothesize that Metformin will protect DM mice from occlusive arterial thrombus formation by altering platelet activation and mitochondrial bioenergetics. Methods DM was developed by low dose of Streptozotocin, healthy (non-DM) mice are controls. Either vehicle or Metformin was administered twice daily via oral gavage for 7-days. Ferric chloride (FeCl3) arterial thrombosis and tail bleeding time were performed. Whole blood aggregometry, platelet activation/adhesion and mitochondrial bioenergetics were evaluated. Results Metformin decreased susceptibility of DM mice to arterial thrombosis. Platelet bioenergetics show DM mice have increased platelet mitochondrial respiration, but no differences were observed with Metformin treatment. In healthy mice, Metformin modulated ADP-dependent increase in platelet adhesion. In healthy mice, Metformin shortens bleeding time with faster thrombotic occlusion. Metformin also increased platelet mitochondrial maximal respiration and spare respiratory capacity uniquely in healthy mice. Conclusion Metformin regulates platelet bioenergetics and ADP-mediated platelet function in DM mice which attenuates susceptibility to arterial thrombosis. Future studies will evaluate clinically relevant doses of Metformin that regulates thrombotic function in diabetic platelets.
Collapse
Affiliation(s)
| | | | | | - Zeyu Liu
- University of Pittsburgh Medical Center
| | | | | | | | | | | |
Collapse
|
6
|
Pankowska KA, Będkowska GE, Chociej-Stypułkowska J, Rusak M, Dąbrowska M, Osada J. Crosstalk of Immune Cells and Platelets in an Ovarian Cancer Microenvironment and Their Prognostic Significance. Int J Mol Sci 2023; 24:ijms24119279. [PMID: 37298230 DOI: 10.3390/ijms24119279] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Ovarian cancer (OC) is one of the deadliest gynecological cancers, largely due to the fast development of metastasis and drug resistance. The immune system is a critical component of the OC tumor microenvironment (TME) and immune cells such as T cells, NK cells, and dendritic cells (DC) play a key role in anti-tumor immunity. However, OC tumor cells are well known for evading immune surveillance by modulating the immune response through various mechanisms. Recruiting immune-suppressive cells such as regulatory T cells (Treg cells), macrophages, or myeloid-derived suppressor cells (MDSC) inhibit the anti-tumor immune response and promote the development and progression of OC. Platelets are also involved in immune evasion by interaction with tumor cells or through the secretion of a variety of growth factors and cytokines to promote tumor growth and angiogenesis. In this review, we discuss the role and contribution of immune cells and platelets in TME. Furthermore, we discuss their potential prognostic significance to help in the early detection of OC and to predict disease outcome.
Collapse
Affiliation(s)
- Katarzyna Aneta Pankowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Grażyna Ewa Będkowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Joanna Chociej-Stypułkowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Małgorzata Rusak
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Milena Dąbrowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Joanna Osada
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| |
Collapse
|
7
|
Filippelli A, Del Gaudio C, Simonis V, Ciccone V, Spini A, Donnini S. Scoping Review on Platelets and Tumor Angiogenesis: Do We Need More Evidence or Better Analysis? Int J Mol Sci 2022; 23:13401. [PMID: 36362186 PMCID: PMC9656254 DOI: 10.3390/ijms232113401] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/28/2022] [Accepted: 10/30/2022] [Indexed: 08/24/2023] Open
Abstract
Platelets are an active component of the tumor microenvironment (TME), involved in the regulation of multiple tumor processes, including angiogenesis. They are generated rich in angiogenic factors in their granules to actively participate in the hemostatic process by megakaryocytes and further enriched in angiogenic factors by all components of the tumor microenvironment to control the angiogenic process because of their preferential relationship with the endothelial component of vessels. In recent decades, the literature has reported a great deal of evidence on the role of platelets in tumor angiogenesis; however, it is unclear whether the number or mean volume of platelets and/or their content and localization in TME may have clinical relevance in the choice and management of therapy for the cancer patient. In this scoping review, we collected and critically reviewed the scientific evidence supporting a close relationship between platelets, cancer, and angiogenesis. The aim of this work was to define the landscape of platelet-activated angiogenesis in cancer progression and analyze what and how much evidence is present in the last 20 years in the literature at both the preclinical and clinical levels, to answer whether platelets could be a useful determinant for analyzing tumor angiogenesis. In conclusion, this scoping review indicates that there is much evidence, both preclinical and clinical, but in the preclinical context, studies demonstrate the direct involvement of platelets in tumor angiogenesis; in the clinical context the evidence is indirect, though strong, and the indication of how and to what extent platelet content contributes to tumor angiogenesis is lacking. So, do we need more evidence or better analysis? More molecular and quali-quantitative data is needed to translate the results obtained in preclinical studies into the clinical setting. This information about platelets, if correlated with tumor type and its biology, including tumor vasculature, type of angiogenesis, and patient characteristics (age, sex, comorbidities, drug treatments for chronic diseases) could be an important pa- rameter for correlating platelet biology to angiogenesis, for personalizing cancer therapy, and for clinical prognosis.
Collapse
Affiliation(s)
- Arianna Filippelli
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Cinzia Del Gaudio
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Vittoria Simonis
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Valerio Ciccone
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Andrea Spini
- Department of Medical Science, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
- Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy
| | - Sandra Donnini
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| |
Collapse
|
8
|
Metformin Mitigated Obesity-Driven Cancer Aggressiveness in Tumor-Bearing Mice. Int J Mol Sci 2022; 23:ijms23169134. [PMID: 36012397 PMCID: PMC9408975 DOI: 10.3390/ijms23169134] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/09/2022] [Accepted: 08/13/2022] [Indexed: 11/17/2022] Open
Abstract
Metformin may offer benefits to certain cancer populations experiencing metabolic abnormalities. To extend the anticancer studies of metformin, a tumor model was established through the implantation of murine Lewis Lung Carcinoma (LLC) cells to Normal Diet (ND)-fed and High-Fat Diet (HFD)-fed C57BL/6 mice. The HFD-fed mice displayed metabolic and pro-inflammatory alterations together with accompanying aggressive tumor growth. Metformin mitigated tumor growth in HFD-fed mice, paralleled by reductions in circulating glucose, insulin, soluble P-selectin, TGF-β1 and High Mobility Group Box-1 (HMGB1), as well as tumor expression of cell proliferation, aerobic glycolysis, glutaminolysis, platelets and neutrophils molecules. The suppressive effects of metformin on cell proliferation, migration and oncogenic signaling molecules were confirmed in cell study. Moreover, tumor-bearing HFD-fed mice had higher contents of circulating and tumor immunopositivity of Neutrophil Extracellular Traps (NETs)-associated molecules, with a suppressive effect from metformin. Data taken from neutrophil studies confirmed the inhibitory effect that metformin has on NET formation induced by HMGB1. Furthermore, HMGB1 was identified as a promoting molecule to boost the transition process towards NETs. The current study shows that metabolic, pro-inflammatory and NET alterations appear to play roles in the obesity-driven aggressiveness of cancer, while also representing candidate targets for anticancer potential of metformin.
Collapse
|
9
|
Barczyński B, Frąszczak K, Kotarski J. Perspectives of metformin use in endometrial cancer and other gynaecological malignancies. J Drug Target 2021; 30:359-367. [PMID: 34753372 DOI: 10.1080/1061186x.2021.2005072] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Insulin resistance and hyperinsulinemia play a key role in type 1 endometrial cancer pathogenesis. Most of these cancers develop on a background of overweight or type 2 diabetes mellitus (T2DM). One of the medications widely used in the treatment of T2DM is biguanide derivative, metformin, which exerts promising anticancer properties principally through activation of adenosine monophosphate kinase (AMPK) and inhibition of mammalian target of rapamycin (mTOR) pathways. Many epidemiological studies on diabetic patients show potential preventative role of metformin in endometrial cancer patients, but data regarding its therapeutic role is still limited. So far, most of attention has been paid to the concept of metformin use in fertility sparing treatment of early-stage cancer. Another investigated alternative is its application in patients with primary advanced or recurrent disease. In this review we present the latest data on clinical use of metformin in endometrial cancer patients and potential underlying mechanisms of its activity. Finally, we present some most important clinical information regarding metformin efficacy in other gynaecological malignancies, mainly breast and ovarian cancer.
Collapse
Affiliation(s)
- Bartłomiej Barczyński
- Ist Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Lublin, Poland
| | - Karolina Frąszczak
- Ist Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Lublin, Poland
| | - Jan Kotarski
- Ist Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
10
|
Hill CN, Hernández-Cáceres MP, Asencio C, Torres B, Solis B, Owen GI. Deciphering the Role of the Coagulation Cascade and Autophagy in Cancer-Related Thrombosis and Metastasis. Front Oncol 2020; 10:605314. [PMID: 33365273 PMCID: PMC7750537 DOI: 10.3389/fonc.2020.605314] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 10/29/2020] [Indexed: 01/10/2023] Open
Abstract
Thrombotic complications are the second leading cause of death among oncology patients worldwide. Enhanced thrombogenesis has multiple origins and may result from a deregulation of megakaryocyte platelet production in the bone marrow, the synthesis of coagulation factors in the liver, and coagulation factor signaling upon cancer and the tumor microenvironment (TME). While a hypercoagulable state has been attributed to factors such as thrombocytosis, enhanced platelet aggregation and Tissue Factor (TF) expression on cancer cells, further reports have suggested that coagulation factors can enhance metastasis through increased endothelial-cancer cell adhesion and enhanced endothelial cell activation. Autophagy is highly associated with cancer survival as a double-edged sword, as can both inhibit and promote cancer progression. In this review, we shall dissect the crosstalk between the coagulation cascade and autophagic pathway and its possible role in metastasis and cancer-associated thrombosis formation. The signaling of the coagulation cascade through the autophagic pathway within the hematopoietic stem cells, the endothelial cell and the cancer cell are discussed. Relevant to the coagulation cascade, we also examine the role of autophagy-related pathways in cancer treatment. In this review, we aim to bring to light possible new areas of cancer investigation and elucidate strategies for future therapeutic intervention.
Collapse
Affiliation(s)
- Charlotte Nicole Hill
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | | | - Catalina Asencio
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Begoña Torres
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Benjamin Solis
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gareth I Owen
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Santiago, Chile.,Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
11
|
Urpilainen E, Puistola U, Boussios S, Karihtala P. Metformin and ovarian cancer: the evidence. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1711. [PMID: 33490223 PMCID: PMC7812201 DOI: 10.21037/atm-20-1060] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In recent decades, great interest in the off-label use of metformin has arisen as a result of its broad effects on different signaling pathways, with only a few side effects, and low cost. Metformin has been shown to have multiple, dose-dependent preclinical anticancer effects, which can be roughly divided into either direct effects via inhibition of mitochondrial respiratory chain complex I, or indirect effects through lowered glucose, insulin and insulin-like growth factor levels. Further details on in vitro and in vivo anticancer effects specifically in ovarian cancer are continuously reported. Preclinically metformin has clear chemosensitizing effects in ovarian cancer and it is an effective negative regulator of angiogenesis. There are also some epidemiological studies on metformin use in ovarian cancer, but the results of these studies are not as promising as those preclinical studies would indicate. Most preclinical studies have involved metformin concentrations that are many times higher than the pharmacological doses used in patients, which might confound the clinical use of metformin as regards the above-mentioned aspects. In this review we evaluate preclinical and clinical evidence concerning metformin in ovarian cancer treatment.
Collapse
Affiliation(s)
- Elina Urpilainen
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu and University Hospital of Oulu, Oulu, Finland
| | - Ulla Puistola
- Department of Obstetrics and Gynaecology, PEDEGO Research Unit, Medical Research Center Oulu, University of Oulu and University Hospital of Oulu, Oulu, Finland
| | - Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham, Kent, UK.,AELIA Organization, 9th Km Thessaloniki - Thermi, Thessaloniki, Greece
| | - Peeter Karihtala
- Department of Oncology and Radiotherapy, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland.,Department of Oncology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Centre, Helsinki, Finland
| |
Collapse
|
12
|
Boussios S, Pavlidis N. Ovarian cancer: state of the art and perspectives of clinical research. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1702. [PMID: 33490214 PMCID: PMC7812238 DOI: 10.21037/atm-2020-oc-14] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Stergios Boussios
- Department of Medical Oncology, Medway NHS Foundation Trust, Windmill Road, ME7 5NY, Gillingham, Kent, UK.,AELIA Organization, 9th Km Thessaloniki - Thermi, 57001, Thessaloniki, Greece
| | - Nicholas Pavlidis
- Medical School, University of Ioannina, Stavros Niarchou Avenue, 45110, Ioannina, Greece
| |
Collapse
|
13
|
Amable G, Martínez-León E, Picco ME, Nemirovsky SI, Rozengurt E, Rey O. Metformin inhibition of colorectal cancer cell migration is associated with rebuilt adherens junctions and FAK downregulation. J Cell Physiol 2020; 235:8334-8344. [PMID: 32239671 PMCID: PMC7529638 DOI: 10.1002/jcp.29677] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 03/06/2020] [Indexed: 01/04/2023]
Abstract
E-cadherin, a central component of the adherens junction (AJ), is a single-pass transmembrane protein that mediates cell-cell adhesion. The loss of E-cadherin surface expression, and therefore cell-cell adhesion, leads to increased cell migration and invasion. Treatment of colorectal cancer (CRC)-derived cells (SW-480 and HT-29) with 2.0 mM metformin promoted a redistribution of cytosolic E-cadherin to de novo formed puncta along the length of the contacting membranes of these cells. Metformin also promoted translocation from the cytosol to the plasma membrane of p120-catenin, another core component of the AJs. Furthermore, E-cadherin and p120-catenin colocalized with β-catenin at cell-cell contacts. Western blot analysis of lysates of CRC-derived cells revealed a substantial metformin-induced increase in the level of p120-catenin as well as E-cadherin phosphorylation on Ser838/840 , a modification associated with β-catenin/E-cadherin interaction. These modifications in E-cadherin, p120-catenin and β-catenin localization suggest that metformin induces rebuilding of AJs in CRC-derived cells. Those modifications were accompanied by the inhibition of focal adhesion kinase (FAK), as revealed by a significant decrease in the phosphorylation of FAK at Tyr397 and paxillin at Tyr118 . These changes were associated with a reduction in the numbers, but an increase in the size, of focal adhesions and by the inhibition of cell migration. Overall, these observations indicate that metformin targets multiple pathways associated with CRC development and progression.
Collapse
Affiliation(s)
- Gastón Amable
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas “José de San Martín”, Ciudad Autónoma de Buenos Aires, 1120, Argentina
| | - Eduardo Martínez-León
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas “José de San Martín”, Ciudad Autónoma de Buenos Aires, 1120, Argentina
| | - María Elisa Picco
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas “José de San Martín”, Ciudad Autónoma de Buenos Aires, 1120, Argentina
| | - Sergio I. Nemirovsky
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, 1428EGA, Argentina
| | - Enrique Rozengurt
- Unit of Signal Transduction and Gastrointestinal Cancer, Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center and Molecular Biology Institute, David Geffen School of Medicine, University of California at Los Angeles, CA, 90095-1768, USA
| | - Osvaldo Rey
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas “José de San Martín”, Ciudad Autónoma de Buenos Aires, 1120, Argentina
| |
Collapse
|
14
|
Hufnagel DH, Cozzi GD, Crispens MA, Beeghly-Fadiel A. Platelets, Thrombocytosis, and Ovarian Cancer Prognosis: Surveying the Landscape of the Literature. Int J Mol Sci 2020; 21:ijms21218169. [PMID: 33142915 PMCID: PMC7663176 DOI: 10.3390/ijms21218169] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 12/16/2022] Open
Abstract
Platelets are critical components of a number of physiologic processes, including tissue remodeling after injury, wound healing, and maintenance of vascular integrity. Increasing evidence suggests that platelets may also play important roles in cancer. In ovarian cancer, thrombocytosis, both at the time of initial diagnosis and at recurrence, has been associated with poorer prognosis. This review describes current evidence for associations between thrombocytosis and ovarian cancer prognosis and discusses the clinical relevance of platelet count thresholds and timing of assessment. In addition, we discuss several mechanisms from in vitro, in vivo, and clinical studies that may underlie these associations and recommend potential approaches for novel therapeutic targets for this lethal disease.
Collapse
Affiliation(s)
- Demetra H. Hufnagel
- Vanderbilt University School of Medicine, 2209 Garland Avenue, Nashville, TN 37240, USA; (D.H.H.); (G.D.C.)
| | - Gabriella D. Cozzi
- Vanderbilt University School of Medicine, 2209 Garland Avenue, Nashville, TN 37240, USA; (D.H.H.); (G.D.C.)
| | - Marta A. Crispens
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Vanderbilt University Medical Center, 1161 21st Avenue South, Nashville, TN 37232, USA;
- Vanderbilt-Ingram Cancer Center, 1301 Medical Center Drive, Nashville, TN 37232, USA
| | - Alicia Beeghly-Fadiel
- Vanderbilt-Ingram Cancer Center, 1301 Medical Center Drive, Nashville, TN 37232, USA
- Department of Medicine, Division of Epidemiology, Vanderbilt University Medical Center, 2525 West End Avenue, Nashville, TN 37203, USA
- Correspondence:
| |
Collapse
|
15
|
Reprogramming of Mesothelial-Mesenchymal Transition in Chronic Peritoneal Diseases by Estrogen Receptor Modulation and TGF-β1 Inhibition. Int J Mol Sci 2020; 21:ijms21114158. [PMID: 32532126 PMCID: PMC7312018 DOI: 10.3390/ijms21114158] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/31/2020] [Accepted: 06/04/2020] [Indexed: 12/14/2022] Open
Abstract
In chronic peritoneal diseases, mesothelial-mesenchymal transition is determined by cues from the extracellular environment rather than just the cellular genome. The transformation of peritoneal mesothelial cells and other host cells into myofibroblasts is mediated by cell membrane receptors, Transforming Growth Factor β1 (TGF-β1), Src and Hypoxia-inducible factor (HIF). This article provides a narrative review of the reprogramming of mesothelial mesenchymal transition in chronic peritoneal diseases, drawing on the similarities in pathophysiology between encapsulating peritoneal sclerosis and peritoneal metastasis, with a particular focus on TGF-β1 signaling and estrogen receptor modulators. Estrogen receptors act at the cell membrane/cytosol as tyrosine kinases that can phosphorylate Src, in a similar way to other receptor tyrosine kinases; or can activate the estrogen response element via nuclear translocation. Tamoxifen can modulate estrogen membrane receptors, and has been shown to be a potent inhibitor of mesothelial-mesenchymal transition (MMT), peritoneal mesothelial cell migration, stromal fibrosis, and neoangiogenesis in the treatment of encapsulating peritoneal sclerosis, with a known side effect and safety profile. The ability of tamoxifen to inhibit the transduction pathways of TGF-β1 and HIF and achieve a quiescent peritoneal stroma makes it a potential candidate for use in cancer treatments. This is relevant to tumors that spread to the peritoneum, particularly those with mesenchymal phenotypes, such as colorectal CMS4 and MSS/EMT gastric cancers, and pancreatic cancer with its desmoplastic stroma. Morphological changes observed during mesothelial mesenchymal transition can be treated with estrogen receptor modulation and TGF-β1 inhibition, which may enable the regression of encapsulating peritoneal sclerosis and peritoneal metastasis.
Collapse
|
16
|
Boussios S, Mikropoulos C, Samartzis E, Karihtala P, Moschetta M, Sheriff M, Karathanasi A, Sadauskaite A, Rassy E, Pavlidis N. Wise Management of Ovarian Cancer: On the Cutting Edge. J Pers Med 2020; 10:E41. [PMID: 32455595 PMCID: PMC7354604 DOI: 10.3390/jpm10020041] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/13/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the fifth leading cause of cancer mortality among women. Two-thirds of patients present at advanced stage at diagnosis, and the estimated 5 year survival rate is 20-40%. This heterogeneous group of malignancies has distinguishable etiology and molecular biology. Initially, single-gene sequencing was performed to identify germline DNA variations associated with EOC. However, hereditary EOC syndrome can be explained by germline pathogenic variants (gPVs) in several genes. In this regard, next-generation sequencing (NGS) changed clinical diagnostic testing, allowing assessment of multiple genes simultaneously in a faster and cheaper manner than sequential single gene analysis. As we move into the era of personalized medicine, there is evidence that poly (ADP-ribose) polymerase (PARP) inhibitors exploit homologous recombination (HR) deficiency, especially in breast cancer gene 1 and 2 (BRCA1/2) mutation carriers. Furthermore, extensive preclinical data supported the development of aurora kinase (AURK) inhibitors in specific tumor types, including EOC. Their efficacy may be optimized in combination with chemotherapeutic or other molecular agents. The efficacy of metformin in ovarian cancer prevention is under investigation. Certain mutations, such as ARID1A mutations, and alterations in the phosphatidylinositol 3-kinase (PI3K)/AKT/mTOR pathway, which are specific in ovarian clear cell carcinoma (OCCC) and endometrioid ovarian carcinoma (EnOC), may offer additional therapeutic targets in these clinical entities. Malignant ovarian germ cell tumors (MOGCTs) are rare and randomized trials are extremely challenging for the improvement of the existing management and development of novel strategies. This review attempts to offer an overview of the main aspects of ovarian cancer, catapulted from the molecular mechanisms to therapeutic considerations.
Collapse
Affiliation(s)
- Stergios Boussios
- Medway NHS Foundation Trust, Windmill Road, Gillingham, Kent ME7 5NY, UK; (M.S.); (A.K.); (A.S.)
- AELIA Organization, 9th Km Thessaloniki—Thermi, 57001 Thessaloniki, Greece
| | - Christos Mikropoulos
- St Luke’s Cancer Center, Royal Surrey County Hospital, Egerton Rd, Guildford GU2 7XX, UK;
| | - Eleftherios Samartzis
- Division of Gynecology, University Hospital Zurich, Frauenklinikstrasse 10, CH-8091 Zürich, Switzerland;
| | - Peeter Karihtala
- Department of Oncology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, P.O. Box 100, FI-00029 Helsinki, Finland;
| | - Michele Moschetta
- Cambridge University Hospitals NHS Foundation Trust, Hills Rd, Cambridge CB2 0QQ, UK;
| | - Matin Sheriff
- Medway NHS Foundation Trust, Windmill Road, Gillingham, Kent ME7 5NY, UK; (M.S.); (A.K.); (A.S.)
| | - Afroditi Karathanasi
- Medway NHS Foundation Trust, Windmill Road, Gillingham, Kent ME7 5NY, UK; (M.S.); (A.K.); (A.S.)
| | - Agne Sadauskaite
- Medway NHS Foundation Trust, Windmill Road, Gillingham, Kent ME7 5NY, UK; (M.S.); (A.K.); (A.S.)
| | - Elie Rassy
- Department of Cancer Medicine, Gustave Roussy Institut, 94805 Villejuif, France;
- Department of Hematology-Oncology, Hotel Dieu de France University Hospital, Faculty of Medicine, Saint Joseph University, Beirut 166830, Lebanon
| | - Nicholas Pavlidis
- Medical School, University of Ioannina, Stavros Niarchou Avenue, 45110 Ioannina, Greece;
| |
Collapse
|
17
|
Fernando K, Yang HW, Jiang Y, Jeon YJ, Ryu B. Ishige okamurae Extract and Its Constituent Ishophloroglucin A Attenuated In Vitro and In Vivo High Glucose-Induced Angiogenesis. Int J Mol Sci 2019; 20:E5542. [PMID: 31698871 PMCID: PMC6888214 DOI: 10.3390/ijms20225542] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 10/29/2019] [Accepted: 11/04/2019] [Indexed: 12/12/2022] Open
Abstract
Diabetes is associated with vascular complications, such as impaired wound healing and accelerated vascular growth. The different clinical manifestations, such as retinopathy and nephropathy, reveal the severity of enhanced vascular growth known as angiogenesis. This study was performed to evaluate the effects of an extract of Ishige okamurae (IO) and its constituent, Ishophloroglucin A (IPA) on high glucose-induced angiogenesis. A transgenic zebrafish (flk:EGFP) embryo model was used to evaluate vessel growth. The 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), gap closure, transwell, and Matrigel® assays were used to analyze the proliferation, migration, and capillary formation of EA.hy926 cells. Moreover, protein expression were determined using western blotting. IO extract and IPA suppressed vessel formation in the transgenic zebrafish (flk:EGFP) embryo. IPA attenuated cell proliferation, cell migration, and capillary-like structure formation in high glucose-treated human vascular endothelial cells. Further, IPA down regulated the expression of high glucose-induced vascular endothelial growth factor receptor 2 (VEGFR-2) and downstream signaling molecule cascade. Overall, the IO extract and IPA exhibited anti-angiogenic effects against high glucose-induced angiogenesis, suggesting their potential for use as therapeutic agents in diabetes-related angiogenesis.
Collapse
Affiliation(s)
- K.H.N. Fernando
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Korea (H.-W.Y.); (Y.J.)
| | - Hye-Won Yang
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Korea (H.-W.Y.); (Y.J.)
| | - Yunfei Jiang
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Korea (H.-W.Y.); (Y.J.)
| | - You-Jin Jeon
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Korea (H.-W.Y.); (Y.J.)
- Marine Science Institute, Jeju National University, Jeju 63333, Korea
| | - BoMi Ryu
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, Jeju 63243, Korea (H.-W.Y.); (Y.J.)
- Marine Science Institute, Jeju National University, Jeju 63333, Korea
| |
Collapse
|
18
|
Arce M, Pinto MP, Galleguillos M, Muñoz C, Lange S, Ramirez C, Erices R, Gonzalez P, Velasquez E, Tempio F, Lopez MN, Salazar-Onfray F, Cautivo K, Kalergis AM, Cruz S, Lladser Á, Lobos-González L, Valenzuela G, Olivares N, Sáez C, Koning T, Sánchez FA, Fuenzalida P, Godoy A, Contreras Orellana P, Leyton L, Lugano R, Dimberg A, Quest AFG, Owen GI. Coagulation Factor Xa Promotes Solid Tumor Growth, Experimental Metastasis and Endothelial Cell Activation. Cancers (Basel) 2019; 11:cancers11081103. [PMID: 31382462 PMCID: PMC6721564 DOI: 10.3390/cancers11081103] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/11/2019] [Accepted: 07/27/2019] [Indexed: 02/06/2023] Open
Abstract
Hypercoagulable state is linked to cancer progression; however, the precise role of the coagulation cascade is poorly described. Herein, we examined the contribution of a hypercoagulative state through the administration of intravenous Coagulation Factor Xa (FXa), on the growth of solid human tumors and the experimental metastasis of the B16F10 melanoma in mouse models. FXa increased solid tumor volume and lung, liver, kidney and lymph node metastasis of tail-vein injected B16F10 cells. Concentrating on the metastasis model, upon coadministration of the anticoagulant Dalteparin, lung metastasis was significantly reduced, and no metastasis was observed in other organs. FXa did not directly alter proliferation, migration or invasion of cancer cells in vitro. Alternatively, FXa upon endothelial cells promoted cytoskeleton contraction, disrupted membrane VE-Cadherin pattern, heightened endothelial-hyperpermeability, increased inflammatory adhesion molecules and enhanced B16F10 adhesion under flow conditions. Microarray analysis of endothelial cells treated with FXa demonstrated elevated expression of inflammatory transcripts. Accordingly, FXa treatment increased immune cell infiltration in mouse lungs, an effect reduced by dalteparin. Taken together, our results suggest that FXa increases B16F10 metastasis via endothelial cell activation and enhanced cancer cell-endothelium adhesion advocating that the coagulation system is not merely a bystander in the process of cancer metastasis.
Collapse
Affiliation(s)
- Maximiliano Arce
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
| | - Mauricio P Pinto
- Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Macarena Galleguillos
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Catalina Muñoz
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Soledad Lange
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Carolina Ramirez
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Rafaela Erices
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Vicerrectoría de Investigación, Universidad Mayor, Santiago 7510041, Chile
| | - Pamela Gonzalez
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Ethel Velasquez
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Comisión Chilena de Energía Nuclear (CCHEN), Santiago, Chile
| | - Fabián Tempio
- Institute of Biomedical Sciences, Faculty of Medicine, University de Chile, Santiago 8380453, Chile
| | - Mercedes N Lopez
- Institute of Biomedical Sciences, Faculty of Medicine, University de Chile, Santiago 8380453, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
| | - Flavio Salazar-Onfray
- Institute of Biomedical Sciences, Faculty of Medicine, University de Chile, Santiago 8380453, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
| | - Kelly Cautivo
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Alexis M Kalergis
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
- Biomedical Research Consortium of Chile, Santiago 8331010, Chile
| | - Sebastián Cruz
- Laboratory of Immunoncology, Fundación Ciencia & Vida, Santiago, Chile
| | - Álvaro Lladser
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile
- Laboratory of Immunoncology, Fundación Ciencia & Vida, Santiago, Chile
| | - Lorena Lobos-González
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Immunoncology, Fundación Ciencia & Vida, Santiago, Chile
- Regenerative Medicine Center, Faculty of Medicine, Clinica Alemana-Universidad Del Desarrollo, Santiago 7650568, Chile
| | - Guillermo Valenzuela
- Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Nixa Olivares
- Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Claudia Sáez
- Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Tania Koning
- Immunology Institute, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Fabiola A Sánchez
- Immunology Institute, Faculty of Medicine, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Patricia Fuenzalida
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Alejandro Godoy
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14203, USA
| | - Pamela Contreras Orellana
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Cellular Communication, ICBM, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Lisette Leyton
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Cellular Communication, ICBM, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Roberta Lugano
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 751 85 Uppsala, Sweden
| | - Andrew F G Quest
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile
- Laboratory of Cellular Communication, ICBM, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Gareth I Owen
- Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile.
- Advanced Center for Chronic Diseases (ACCDiS), Santiago, Chile.
- Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile.
- Millennium Institute on Immunology and Immunotherapy, Santiago 8331150, Chile.
| |
Collapse
|
19
|
Metformin induces the AP-1 transcription factor network in normal dermal fibroblasts. Sci Rep 2019; 9:5369. [PMID: 30926854 PMCID: PMC6441003 DOI: 10.1038/s41598-019-41839-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 03/18/2019] [Indexed: 12/21/2022] Open
Abstract
Metformin is a widely-used treatment for type 2 diabetes and is reported to extend health and lifespan as a caloric restriction (CR) mimetic. Although the benefits of metformin are well documented, the impact of this compound on the function and organization of the genome in normal tissues is unclear. To explore this impact, primary human fibroblasts were treated in culture with metformin resulting in a significant decrease in cell proliferation without evidence of cell death. Furthermore, metformin induced repositioning of chromosomes 10 and 18 within the nuclear volume indicating altered genome organization. Transcriptome analyses from RNA sequencing datasets revealed that alteration in growth profiles and chromosome positioning occurred concomitantly with changes in gene expression profiles. We further identified that different concentrations of metformin induced different transcript profiles; however, significant enrichment in the activator protein 1 (AP-1) transcription factor network was common between the different treatments. Comparative analyses revealed that metformin induced divergent changes in the transcriptome than that of rapamycin, another proposed mimetic of CR. Promoter analysis and chromatin immunoprecipitation assays of genes that changed expression in response to metformin revealed enrichment of the transcriptional regulator forkhead box O3a (FOXO3a) in normal human fibroblasts, but not of the predicted serum response factor (SRF). Therefore, we have demonstrated that metformin has significant impacts on genome organization and function in normal human fibroblasts, different from those of rapamycin, with FOXO3a likely playing a role in this response.
Collapse
|
20
|
Fernando KHN, Yang HW, Jiang Y, Jeon YJ, Ryu B. Diphlorethohydroxycarmalol Isolated from Ishige okamurae Represses High Glucose-Induced Angiogenesis In Vitro and In Vivo. Mar Drugs 2018; 16:E375. [PMID: 30308943 PMCID: PMC6215322 DOI: 10.3390/md16100375] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 10/04/2018] [Accepted: 10/05/2018] [Indexed: 12/12/2022] Open
Abstract
Diabetes mellitus causes abnormalities of angiogenesis leading to vascular dysfunction and serious pathologies. Diphlorethohydroxycarmalol (DPHC), which is isolated from Ishige okamurae, is well known for its bioactivities, including antihyperglycemic and protective functions against diabetes-related pathologies. In the present study, the inhibitory effect of DPHC on high glucose-induced angiogenesis was investigated on the human vascular endothelial cell line EA.hy926. DPHC inhibited the cell proliferation, cell migration, and tube formation in cells exposed to 30 mM of glucose to induce angiogenesis. Furthermore, the effect of DPHC against high glucose-induced angiogenesis was evaluated in zebrafish embryos. The treatment of embryos with DPHC suppressed high glucose-induced dilation in the retinal vessel diameter and vessel formation. Moreover, DPHC could inhibit high glucose-induced vascular endothelial growth factor receptor 2 (VEGFR-2) expression and its downstream signaling cascade. Overall, these findings suggest that DPHC is actively involved in the suppression of high glucose-induced angiogenesis. Hence, DPHC is a potential agent for the development of therapeutics against angiogenesis induced by diabetes.
Collapse
Affiliation(s)
- K H N Fernando
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea.
| | - Hye-Won Yang
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea.
| | - Yunfei Jiang
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea.
| | - You-Jin Jeon
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea.
| | - BoMi Ryu
- Department of Marine Life Science, Jeju National University, Jeju 63243, Korea.
| |
Collapse
|
21
|
Wilson RB. Hypoxia, cytokines and stromal recruitment: parallels between pathophysiology of encapsulating peritoneal sclerosis, endometriosis and peritoneal metastasis. Pleura Peritoneum 2018; 3:20180103. [PMID: 30911653 PMCID: PMC6405013 DOI: 10.1515/pp-2018-0103] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 03/01/2018] [Indexed: 02/06/2023] Open
Abstract
Peritoneal response to various kinds of injury involves loss of peritoneal mesothelial cells (PMC), danger signalling, epithelial-mesenchymal transition and mesothelial-mesenchymal transition (MMT). Encapsulating peritoneal sclerosis (EPS), endometriosis (EM) and peritoneal metastasis (PM) are all characterized by hypoxia and formation of a vascularized connective tissue stroma mediated by vascular endothelial growth factor (VEGF). Transforming growth factor-β1 (TGF-β1) is constitutively expressed by the PMC and plays a major role in the maintenance of a transformed, inflammatory micro-environment in PM, but also in EPS and EM. Persistently high levels of TGF-β1 or stimulation by inflammatory cytokines (interleukin-6 (IL-6)) induce peritoneal MMT, adhesion formation and fibrosis. TGF-β1 enhances hypoxia inducible factor-1α expression, which drives cell growth, extracellular matrix production and cell migration. Disruption of the peritoneal glycocalyx and exposure of the basement membrane release low molecular weight hyaluronan, which initiates a cascade of pro-inflammatory mediators, including peritoneal cytokines (TNF-α, IL-1, IL-6, prostaglandins), growth factors (TGF-α, TGF-β, platelet-derived growth factor, VEGF, epidermal growth factor) and the fibrin/coagulation cascade (thrombin, Tissue factor, plasminogen activator inhibitor [PAI]-1/2). Chronic inflammation and cellular transformation are mediated by damage-associated molecular patterns, pattern recognition receptors, AGE-RAGE, extracellular lactate, pro-inflammatory cytokines, reactive oxygen species, increased glycolysis, metabolomic reprogramming and cancer-associated fibroblasts. The pathogenesis of EPS, EM and PM shows similarities to the cellular transformation and stromal recruitment of wound healing.
Collapse
Affiliation(s)
- Robert Beaumont Wilson
- Upper GI Surgery Department, Liverpool Hospital, Elizabeth St, Liverpool, 2170, NSW, Australia
| |
Collapse
|
22
|
Cuello MA, Kato S, Liberona F. The impact on high-grade serous ovarian cancer of obesity and lipid metabolism-related gene expression patterns: the underestimated driving force affecting prognosis. J Cell Mol Med 2017; 22:1805-1815. [PMID: 29266765 PMCID: PMC5824367 DOI: 10.1111/jcmm.13463] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 10/21/2017] [Indexed: 01/06/2023] Open
Abstract
To investigate whether specific obesity/metabolism‐related gene expression patterns affect the survival of patients with ovarian cancer. Clinical and genomic data of 590 samples from the high‐grade ovarian serous carcinoma (HGOSC) study of The Cancer Genome Atlas (TCGA) and 91 samples from the Australian Ovarian Cancer Study were downloaded from the International Cancer Genome Consortium (ICGC) portal. Clustering of mRNA microarray and reverse‐phase protein array (RPPA) data was performed with 83 consensus driver genes and 144 obesity and lipid metabolism‐related genes. Association between different clusters and survival was analyzed with the Kaplan–Meier method and a Cox regression. Mutually exclusive, co‐occurrence and network analyses were also carried out. Using RNA and RPPA data, it was possible to identify two subsets of HGOSCs with similar clinical characteristics and cancer driver mutation profiles (e.g. TP53), but with different outcome. These differences depend more on up‐regulation of specific obesity and lipid metabolism‐related genes than on the number of gene mutations or copy number alterations. It was also found that CD36 and TGF‐ß are highly up‐regulated at the protein levels in the cluster with the poorer outcome. In contrast, BSCL2 is highly up‐regulated in the cluster with better progression‐free and overall survival. Different obesity/metabolism‐related gene expression patterns constitute a risk factor for prognosis independent of the therapy results in the Cox regression. Prognoses were conditioned by the differential expression of obesity and lipid metabolism‐related genes in HGOSCs with similar cancer driver mutation profiles, independent of the initial therapeutic response.
Collapse
Affiliation(s)
- Mauricio A Cuello
- Division of Obstetrics and Gynecology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sumie Kato
- Division of Obstetrics and Gynecology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisca Liberona
- Division of Obstetrics and Gynecology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
23
|
Papanagnou P, Stivarou T, Papageorgiou I, Papadopoulos GE, Pappas A. Marketed drugs used for the management of hypercholesterolemia as anticancer armament. Onco Targets Ther 2017; 10:4393-4411. [PMID: 28932124 PMCID: PMC5598753 DOI: 10.2147/ott.s140483] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The design of novel pharmacologic agents as well as their approval for sale in markets all over the world is a tedious and pricey process. Inevitably, oncologic patients commonly experience unwanted effects of new anticancer drugs, while the acquisition of clinical experience for these drugs is largely based on doctor–patient partnership which is not always effective. The repositioning of marketed non-antineoplastic drugs that hopefully exhibit anticancer properties into the field of oncology is a challenging option that gains ground and attracts preclinical and clinical research in an effort to override all these hindrances and minimize the risk for reduced efficacy and/or personalized toxicity. This review aims to present the anticancer properties of drugs used for the management of hypercholesterolemia. A global view of the antitumorigenicity of all marketed antihypercholesterolemic drugs is of major importance, given that atherosclerosis, which is etiologically linked to hypercholesterolemia, is a leading worldwide cause of morbidity and mortality, while hypercholesterolemia and tumorigenesis are known to be interrelated. In vitro, in vivo and clinical literature data accumulated so far outline the mechanistic basis of the antitumor function of these agents and how they could find application at the clinical setting.
Collapse
Affiliation(s)
| | - Theodora Stivarou
- Immunology Laboratory, Immunology Department, Hellenic Pasteur Institute, Athens, Greece
| | | | | | | |
Collapse
|