1
|
Faria PCL, Resende RS, Cardoso AM. Metastasis and angiogenesis in cervical cancer: key aspects of purinergic signaling in platelets and possible therapeutic targets. Purinergic Signal 2024; 20:607-616. [PMID: 38753131 PMCID: PMC11554953 DOI: 10.1007/s11302-024-10020-3] [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: 09/28/2023] [Accepted: 05/09/2024] [Indexed: 11/13/2024] Open
Abstract
Cervical cancer ranks as the fourth most common and fatal cancer among women worldwide. Studies have demonstrated a strong association between purinergic platelet signaling and tumor progression in this type of cancer. The literature shows that neoplastic cells, when in the bloodstream, secrete adenosine triphosphate (ATP) and adenosine nucleotide diphosphate (ADP) that act on their corresponding platelet P2Y and P2X receptors. The interaction of these nucleotides with their receptors results in platelet activation and degranulation, ensuing several consequences, such as vascular endothelial growth factor (VEGF), platelet-derived growth factor, matrix metalloproteinases, ADP, and ATP. These molecules play essential roles in angiogenesis and tumor metastasis in cervical cancer. Several purinergic receptors are found in endothelial cells. Their activation, especially P2Y2, by the nucleotides released by platelets can induce relaxation of the endothelial barrier and consequent extravasation of tumor cells, promoting the development of metastases. Cancer cells that enter the bloodstream during the metastatic process are also subject to high shear stress and immune surveillance. In this context, activated platelets bind to circulating tumor cells and protect them against shear stress and the host's immune system, especially against natural killer cells, facilitating their spread throughout the body. Furthermore, activation of the P2Y12 receptor present on the platelet surface promotes the release of VEGF, the main inducer of angiogenesis in cervical cancer, in addition to increasing the concentration of several other pro-angiogenic molecules. Therefore, this review will address the role of platelet purinergic signaling in tumor progression of cervical cancer and propose possible therapeutic targets.
Collapse
Affiliation(s)
- Paula C L Faria
- Medical School, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Rackel S Resende
- Medical School, Federal University of Fronteira Sul, Chapecó, SC, Brazil
| | - Andréia M Cardoso
- Graduate Program in Biomedical Sciences, Federal University of Fronteira Sul, Chapecó, SC, Brazil.
| |
Collapse
|
2
|
Souza JLN, Antunes-Porto AR, da Silva Oliveira I, Amorim CCO, Pires LO, de Brito Duval I, Amaral LVBD, Souza FR, Oliveira EA, Cassali GD, Cardoso VN, Fernandes SOA, Fujiwara RT, Russo RC, Bueno LL. Screening and validating the optimal panel of housekeeping genes for 4T1 breast carcinoma and metastasis studies in mice. Sci Rep 2024; 14:26476. [PMID: 39488625 PMCID: PMC11531515 DOI: 10.1038/s41598-024-77126-x] [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: 05/17/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024] Open
Abstract
The 4T1 model is extensively employed in murine studies to elucidate the mechanisms underlying the carcinogenesis of triple-negative breast cancer. Molecular biology serves as a cornerstone in these investigations. However, accurate gene expression analyses necessitate data normalization employing housekeeping genes (HKGs) to avert spurious results. Here, we initially delve into the characteristics of the tumor evolution induced by 4T1 in mice, underscoring the imperative for additional tools for tumor monitoring and assessment methods for tracking the animals, thereby facilitating prospective studies employing this methodology. Subsequently, leveraging various software platforms, we assessed ten distinct HKGs (GAPDH, 18 S, ACTB, HPRT1, B2M, GUSB, PGK1, CCSER2, SYMPK, ANKRD17) not hitherto evaluated in the 4T1 breast cancer model, across tumors and diverse tissues afflicted by metastasis. Our principal findings underscore GAPDH as the optimal HKG for gene expression analyses in tumors, while HPRT1 emerged as the most stable in the liver and CCSER2 in the lung. These genes demonstrated consistent expression and minimal variation among experimental groups. Furthermore, employing these HKGs for normalization, we assessed TNF-α and VEGF expression in tissues and discerned significant disparities among groups. We posit that this constitutes the inaugural delineation of an ideal HKG for experiments utilizing the 4T1 model, particularly in vivo settings.
Collapse
Affiliation(s)
- Jorge Lucas Nascimento Souza
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Rafaela Antunes-Porto
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Izabela da Silva Oliveira
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Chiara Cássia Oliveira Amorim
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiz Octávio Pires
- Laboratory of Radioisotopes, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Isabela de Brito Duval
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luisa Vitor Braga do Amaral
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda Rezende Souza
- Laboratory of Comparative Pathology, Department of Genetal Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Evelyn Ane Oliveira
- Laboratory of Comparative Pathology, Department of Genetal Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Geovanni Dantas Cassali
- Laboratory of Comparative Pathology, Department of Genetal Pathology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Valbert Nascimento Cardoso
- Laboratory of Radioisotopes, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Simone Odília Antunes Fernandes
- Laboratory of Radioisotopes, Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Toshio Fujiwara
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lilian Lacerda Bueno
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
- Laboratory of Immunobiology and Control of Parasites, Department of Parasitology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, 31270- 901, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
3
|
Kast RE. IC Regimen: Delaying Resistance to Lorlatinib in ALK Driven Cancers by Adding Repurposed Itraconazole and Cilostazol. Cells 2024; 13:1175. [PMID: 39056757 PMCID: PMC11274432 DOI: 10.3390/cells13141175] [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: 06/21/2024] [Revised: 07/06/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Lorlatinib is a pharmaceutical ALK kinase inhibitor used to treat ALK driven non-small cell lung cancers. This paper analyses the intersection of past published data on the physiological consequences of two unrelated drugs from general medical practice-itraconazole and cilostazol-with the pathophysiology of ALK positive non-small cell lung cancer. A conclusion from that data analysis is that adding itraconazole and cilostazol may make lorlatinib more effective. Itraconazole, although marketed worldwide as a generic antifungal drug, also inhibits Hedgehog signaling, Wnt signaling, hepatic CYP3A4, and the p-gp efflux pump. Cilostazol, marketed worldwide as a generic thrombosis preventative drug, acts by inhibiting phosphodiesterase 3, and, by so doing, lowers platelets' adhesion, thereby partially depriving malignant cells of the many tumor trophic growth factors supplied by platelets. Itraconazole may enhance lorlatinib effectiveness by (i) reducing or stopping a Hedgehog-ALK amplifying feedback loop, by (ii) increasing lorlatinib's brain levels by p-gp inhibition, and by (iii) inhibiting growth drive from Wnt signaling. Cilostazol, surprisingly, carries minimal bleeding risk, lower than that of aspirin. Risk/benefit assessment of the combination of metastatic ALK positive lung cancer being a low-survival disease with the predicted safety of itraconazole-cilostazol augmentation of lorlatinib favors a trial of this drug trio in ALK positive lung cancer.
Collapse
|
4
|
Lee SB, Ji HD, Lee IK, Kim KS, Lee J, Lee SW, Jeon YH. Visualization of platelet recruitment to tumor lesions using highly sensitive and stable radioiodine studded gold nanoprobes. J Mater Chem B 2021; 9:2931-2936. [PMID: 33885648 DOI: 10.1039/d0tb02265a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
In vivo imaging of platelets will provide a better understanding of their critical roles in arterial cardiovascular disease, hemostasis, inflammation, and cancer. Here, we demonstrate the feasibility of using radioiodine studded gold nanoprobes (RIS-GNPs) as a platelet tracker for nuclear medicine imaging in tumor-bearing mice using positron emission tomography and computed tomography (PET/CT). Platelet labeling with RIS-GNPs did not alter the platelet functions, such as cellular proliferation and aggregation. PET/CT imaging clearly revealed the migration of platelets into tumor sites at 1 to 5 h post-transfer of RIS-GNP-labeled platelets, which was consistent with the biodistribution data. Our findings suggest that the imaging approach using RIS-GNPs makes it feasible to visualize the biological behavior of platelets in living organisms with cancer.
Collapse
Affiliation(s)
- Sang Bong Lee
- Vaccine Commercialization Center, Gyeongbuk Institute for Bio industry, 88 Saneopdanji-gil, pungsan-eup, Andong-si, Gyeongbuk, 33618, South Korea
| | | | | | | | | | | | | |
Collapse
|
5
|
Ishibashi Y, Tsujimoto H, Kouzu K, Itazaki Y, Tsuchiya S, Fujishima S, Yaguchi Y, Sugasawa H, Nomura S, Ito N, Harada M, Nagata H, Shinto E, Kishi Y, Ueno H. Impact of Antiplatelet and Anticoagulant Therapies on Platelet-related Prognostic Markers in Patients With Esophageal Cancer. In Vivo 2020; 34:1941-1949. [PMID: 32606166 PMCID: PMC7439896 DOI: 10.21873/invivo.11991] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/08/2020] [Accepted: 04/10/2020] [Indexed: 01/17/2023]
Abstract
BACKGROUND/AIM In recent years, platelet-related markers were recognized as useful prognostic factors in various malignancies. We investigated the relationship between platelet-related prognostic markers and anti-platelet or anti-coagulant therapies for survival outcomes in esophageal squamous cell carcinoma. PATIENTS AND METHODS Preoperative platelet-related prognostic markers were evaluated from peripheral blood testing and statistical analyses were performed to evaluate the prognostic value of these markers and reveal the effects of antiplatelets and/or anticoagulants regarding their prognostic relevance. RESULTS In all 176 patients, preoperative platelet-to-lymphocyte ratio (PLR) was not found to be a predictor of overall survival (OS). However, in patients without antiplatelet or anticoagulant therapies, PLR was significantly associated with a poor OS (p=0.03). Although platelet large cell ratio (P-LCR) was not associated with the prognosis in patients with antiplatelet and/or anticoagulant therapies, higher P-LCR was associated with a poor prognosis in patients without antiplatelet or anticoagulant therapies (p<0.0001). CONCLUSION Researching detailed antiplatelet and anticoagulant therapies could reinforce the prognostic value of platelet-related prognostic markers in ESCC.
Collapse
Affiliation(s)
- Yusuke Ishibashi
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | | | - Keita Kouzu
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Yujiro Itazaki
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Satoshi Tsuchiya
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | | | - Yoshihisa Yaguchi
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Hidekazu Sugasawa
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Shinsuke Nomura
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Nozomi Ito
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Manabu Harada
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Hiromi Nagata
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Eiji Shinto
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Yoji Kishi
- Department of Surgery, National Defense Medical College, Saitama, Japan
| | - Hideki Ueno
- Department of Surgery, National Defense Medical College, Saitama, Japan
| |
Collapse
|
6
|
Kassassir H, Karolczak K, Siewiera KM, Wojkowska DW, Braun M, Watala CW. Time-dependent interactions of blood platelets and cancer cells, accompanied by extramedullary hematopoiesis, lead to increased platelet activation and reactivity in a mouse orthotopic model of breast cancer - implications for pulmonary and liver metastasis. Aging (Albany NY) 2020; 12:5091-5120. [PMID: 32191918 PMCID: PMC7138580 DOI: 10.18632/aging.102933] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 02/08/2020] [Indexed: 12/30/2022]
Abstract
Aging has become a significant risk factor for several diseases, including breast cancer. Platelet activation and platelet-cancer cell aggregate fractions were found to increase with tumor progression in a mouse model of breast cancer. At advanced stages of tumor development, platelets from mice with breast cancer were hyperreactive to low agonist concentrations and hyporeactive to high ones. Platelet activation and reactivity were strongly associated with breast cancer metastasis in the lungs and extramedullary hematopoiesis in the liver. A greater fraction of platelet aggregates was observed in 4T1-injected mice at the advanced stages of breast cancer. In vitro, platelet activation was elevated after incubation with 4T1 cells, and thrombin-stimulated platelets formed aggregates with 4T1 cells. Neither GPIbα, nor GPIIb/IIIa blocking antibodies, were able to affect platelet-cancer cell aggregation in vitro. The primed circulating platelets became more sensitive to subthreshold stimuli at advanced stages of tumor development, and the formation of platelet-cancer cell aggregates increased with cancer progression. Our findings demonstrate that the age-associated progression of breast cancer cells is connected with increased platelet functioning, and that it can be manifested by the increased number of metastases and extramedullary hematopoiesis in a time-dependent-manner.
Collapse
Affiliation(s)
- Hassan Kassassir
- Department of Haemostatic Disorders, Faculty of Health Sciences, Medical University of Lodz, Lodz, Poland
| | - Kamil Karolczak
- Department of Haemostatic Disorders, Faculty of Health Sciences, Medical University of Lodz, Lodz, Poland
| | - Karolina M Siewiera
- Department of Haemostatic Disorders, Faculty of Health Sciences, Medical University of Lodz, Lodz, Poland.,Department of Cytobiology and Proteomics, Medical University of Lodz, Lodz, Poland
| | - Dagmara W Wojkowska
- Department of Haemostatic Disorders, Faculty of Health Sciences, Medical University of Lodz, Lodz, Poland
| | - Marcin Braun
- Department of Pathology, Medical University of Lodz, Lodz, Poland.,Postgraduate School of Molecular Medicine, Warsaw Medical University, Warsaw, Poland
| | - Cezary W Watala
- Department of Haemostatic Disorders, Faculty of Health Sciences, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
7
|
Hao N, Shen W, Du R, Jiang S, Zhu J, Chen Y, Huang C, Shi Y, Xiang R, Luo Y. Phosphodiesterase 3A Represents a Therapeutic Target that Drives Stem Cell–like Property and Metastasis in Breast Cancer. Mol Cancer Ther 2019; 19:868-881. [PMID: 31871268 DOI: 10.1158/1535-7163.mct-18-1233] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 02/19/2019] [Accepted: 12/03/2019] [Indexed: 11/16/2022]
MESH Headings
- Animals
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/drug therapy
- Breast Neoplasms/enzymology
- Breast Neoplasms/pathology
- Breast Neoplasms/secondary
- Cell Proliferation
- Cilostazol/pharmacology
- Cyclic Nucleotide Phosphodiesterases, Type 3/chemistry
- Cyclic Nucleotide Phosphodiesterases, Type 3/genetics
- Cyclic Nucleotide Phosphodiesterases, Type 3/metabolism
- Female
- Gene Expression Regulation, Enzymologic/drug effects
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Microfilament Proteins/metabolism
- Middle Aged
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/enzymology
- Neoplastic Stem Cells/metabolism
- Phosphodiesterase 3 Inhibitors/pharmacology
- Prognosis
- Protein Transport
- Signal Transduction
- Tumor Cells, Cultured
- Vesicular Transport Proteins/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Na Hao
- Department of Breast Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
- Department of Immunology, Institute of Basic Medical Science, Chinese Academy of Medical Science, School of Basic Medicine Peking Union Medical College, Beijing, China
- International Joint Center for Biomedical Research of the Ministry of Education, Tianjin, China
| | - Wenzhi Shen
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China
- Department of Immunology, Institute of Basic Medical Science, Chinese Academy of Medical Science, School of Basic Medicine Peking Union Medical College, Beijing, China
- Department of Pathology and Institute of Precision Medicine, Jining Medical University, Jining, China
| | - Renle Du
- Department of Breast Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shan Jiang
- Department of Breast Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Junyong Zhu
- Department of Galactophore, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Yanan Chen
- Department of Breast Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Immunology, Institute of Basic Medical Science, Chinese Academy of Medical Science, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Chongbiao Huang
- Senior Ward, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research, Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yi Shi
- Department of Breast Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Immunology, Institute of Basic Medical Science, Chinese Academy of Medical Science, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Rong Xiang
- Department of Breast Surgery, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Immunology, Institute of Basic Medical Science, Chinese Academy of Medical Science, School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Yunping Luo
- Department of Immunology, School of Medicine, Nankai University, Tianjin, China.
| |
Collapse
|
8
|
Mohamad NE, Yeap SK, Abu N, Lim KL, Zamberi NR, Nordin N, Sharifuddin SA, Long K, Alitheen NB. In vitro and in vivo antitumour effects of coconut water vinegar on 4T1 breast cancer cells. Food Nutr Res 2019; 63:1616. [PMID: 30814922 PMCID: PMC6387426 DOI: 10.29219/fnr.v63.1616] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 09/12/2018] [Accepted: 11/28/2018] [Indexed: 01/01/2023] Open
Abstract
Background Coconut water and vinegars have been reported to possess potential anti-tumour and immunostimulatory effects. However, the anti-tumour, anti-inflammatory and immunostimulatory effects of coconut water vinegar have yet to be tested. Objective This study investigated the in vitro and in vivo anti-tumour effects of coconut water vinegar on 4T1 breast cancer cells. Methods The 4T1 cells were treated with freeze-dried coconut water vinegar and subjected to MTT cell viability, BrdU, annexin V/PI apoptosis, cell cycle and wound healing assays for the in vitro analysis. For the in vivo chemopreventive evaluation, mice challenged with 4T1 cells were treated with 0.08or 2.00 mL/kg body weight of fresh coconut water vinegar for 28 days. Tumour weight, apoptosis of tumour cells, metastasis and immunity of untreated mice and coconut water vinegar-treated 4T1 challenged mice were compared. Results Freeze-dried coconut water vinegar reduced the cell viability, induced apoptosis and delayed the wound healing effect of 4T1 cells in vitro. In vivo, coconut water vinegar delayed 4T1 breast cancer progression in mice by inducing apoptosis and delaying the metastasis. Furthermore, coconut water vinegar also promoted immune cell cytotoxicity and production of anticancer cytokines. The results indicate that coconut water vinegar delays breast cancer progression by inducing apoptosis in breast cancer cells, suppressing metastasis and activating anti-tumour immunity. Conclusion Coconut water vinegar is a potential health food ingredient with a chemopreventive effect.
Collapse
Affiliation(s)
- Nurul Elyani Mohamad
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, Jalan Sunsuria, Bandar Sunsuria, Sepang, Selangor, Malaysia.,Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Nadiah Abu
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.,UKM Medical Centre, UKM Medical Molecular Biology Institute (UMBI), Cheras, Wilayah Persekutuan, Malaysia
| | - Kian Lam Lim
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Jalan Sungai Long, Bandar Sungai Long, Cheras, Kajang, Selangor, Malaysia
| | - Nur Rizi Zamberi
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Noraini Nordin
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Shaiful Adzni Sharifuddin
- Biotechnology Research Centre, Malaysian Agricultural Research and Development Institute (MARDI), Serdang, Selangor, Malaysia
| | - Kamariah Long
- Biotechnology Research Centre, Malaysian Agricultural Research and Development Institute (MARDI), Serdang, Selangor, Malaysia
| | - Noorjahan Banu Alitheen
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia, Serdang, Selangor, Malaysia.,Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
9
|
Yang CH, Hsia CW, Jayakumar T, Sheu JR, Hsia CH, Khamrang T, Chen YJ, Manubolu M, Chang Y. Structure⁻Activity Relationship Study of Newly Synthesized Iridium-III Complexes as Potential Series for Treating Thrombotic Diseases. Int J Mol Sci 2018; 19:ijms19113641. [PMID: 30463221 PMCID: PMC6274890 DOI: 10.3390/ijms19113641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/05/2018] [Accepted: 11/15/2018] [Indexed: 01/07/2023] Open
Abstract
Platelets play a major role in hemostatic events and are associated with various pathological events, such as arterial thrombosis and atherosclerosis. Iridium (Ir) compounds are potential alternatives to platinum compounds, since they exert promising anticancer effects without cellular toxicity. Our recent studies found that Ir compounds show potent antiplatelet properties. In this study, we evaluated the in vitro antiplatelet, in vivo antithrombotic and structure⁻activity relationship (SAR) of newly synthesized Ir complexes, Ir-1, Ir-2 and Ir-4, in agonists-induced human platelets. Among the tested compounds, Ir-1 was active in inhibiting platelet aggregation induced by collagen; however, Ir-2 and Ir-4 had no effects even at their maximum concentrations of 50 μM against collagen and 500 μM against U46619-induced aggregation. Similarly, Ir-1 was potently inhibiting of adenosine triphosphate (ATP) release, calcium mobilization ([Ca2+]i) and P-selectin expression induced by collagen-induced without cytotoxicity. Likewise, Ir-1 expressively suppressed collagen-induced Akt, PKC, p38MAPKs and JNK phosphorylation. Interestingly, Ir-2 and Ir-4 had no effect on platelet function analyzer (PFA-100) collagen-adenosine diphosphate (C-ADP) and collagen-epinephrine (C-EPI) induced closure times in mice, but Ir-1 caused a significant increase when using C-ADP stimulation. Other in vivo studies revealed that Ir-1 significantly prolonged the platelet plug formation, increased tail bleeding times and reduced the mortality of adenosine diphosphate (ADP)-induced acute pulmonary thromboembolism in mice. Ir-1 has no substitution on its phenyl group, a water molecule (like cisplatin) can replace its chloride ion and, hence, the rate of hydrolysis might be tuned by the substituent on the ligand system. These features might have played a role for the observed effects of Ir-1. These results indicate that Ir-1 may be a lead compound to design new antiplatelet drugs for the treatment of thromboembolic diseases.
Collapse
Affiliation(s)
- Chih-Hao Yang
- Department of Pharmacology, Schools of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan.
| | - Chih-Wei Hsia
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan.
| | - Thanasekaran Jayakumar
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan.
| | - Joen-Rong Sheu
- Department of Pharmacology, Schools of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan.
| | - Chih-Hsuan Hsia
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan.
| | - Themmila Khamrang
- Department of Chemistry, North Eastern Hill University, Shillong 793022, India.
| | - Yen-Jen Chen
- Department of Pharmacology, Schools of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan.
| | - Manjunath Manubolu
- Department of Evolution, Ecology and Organismal Biology, Ohio State University, Columbus, OH 43212, USA.
| | - Yi Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan.
- Department of Anesthesiology, Shin Kong Wu Ho-Su Memorial Hospital, No. 95, Wen Chang Rd., Taipei 111, Taiwan.
- School of Medicine, Fu-Jen Catholic University, No. 510, Zhong Zheng Rd, Xin Zhuang Dist., New Taipei City 242, Taiwan.
| |
Collapse
|
10
|
Jayakumar T, Hsu CY, Khamrang T, Hsia CH, Hsia CW, Manubolu M, Sheu JR. Possible Molecular Targets of Novel Ruthenium Complexes in Antiplatelet Therapy. Int J Mol Sci 2018; 19:ijms19061818. [PMID: 29925802 PMCID: PMC6032250 DOI: 10.3390/ijms19061818] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 06/13/2018] [Accepted: 06/15/2018] [Indexed: 12/19/2022] Open
Abstract
In oncotherapy, ruthenium (Ru) complexes are reflected as potential alternatives for platinum compounds and have been proved as encouraging anticancer drugs with high efficacy and low side effects. Cardiovascular diseases (CVDs) are mutually considered as the number one killer globally, and thrombosis is liable for the majority of CVD-related deaths. Platelets, an anuclear and small circulating blood cell, play key roles in hemostasis by inhibiting unnecessary blood loss of vascular damage by making blood clot. Platelet activation also plays a role in cancer metastasis and progression. Nevertheless, abnormal activation of platelets results in thrombosis under pathological settings such as the rupture of atherosclerotic plaques. Thrombosis diminishes the blood supply to the heart and brain resulting in heart attacks and strokes, respectively. While currently used anti-platelet drugs such as aspirin and clopidogrel demonstrate efficacy in many patients, they exert undesirable side effects. Therefore, the development of effective therapeutic strategies for the prevention and treatment of thrombotic diseases is a demanding priority. Recently, precious metal drugs have conquered the subject of metal-based drugs, and several investigators have motivated their attention on the synthesis of various ruthenium (Ru) complexes due to their prospective therapeutic values. Similarly, our recent studies established that novel ruthenium-based compounds suppressed platelet aggregation via inhibiting several signaling cascades. Our study also described the structure antiplatelet-activity relationship (SAR) of three newly synthesized ruthenium-based compounds. This review summarizes the antiplatelet activity of newly synthesized ruthenium-based compounds with their potential molecular mechanisms.
Collapse
Affiliation(s)
- Thanasekaran Jayakumar
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Chia-Yuan Hsu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan.
| | - Themmila Khamrang
- Department of Chemistry, North Eastern Hill University, Shillong 793022, India.
| | - Chih-Hsuan Hsia
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Chih-Wei Hsia
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Manjunath Manubolu
- Department of Evolution, Ecology and Organismal Biology, Ohio State University, Columbus, OH 43212, USA.
| | - Joen-Rong Sheu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
11
|
Abstract
Metastasis contributes to poor prognosis in many types of cancer and is the leading cause of cancer-related deaths. Tumor cells metastasize to distant sites via the circulatory and lymphatic systems. In this review, we discuss the potential of circulating tumor cells for diagnosis and describe the experimental therapeutics that aim to target these disseminating cancer cells. We discuss the advantages and limitations of such strategies and how they may lead to the development of the next generation of antimetastasis treatments.
Collapse
Affiliation(s)
- Eric Lin
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Thong Cao
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, USA
| | - Sunitha Nagrath
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48109, USA
| | - Michael R. King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, USA
| |
Collapse
|
12
|
Ying M, Zhuang J, Wei X, Zhang X, Zhang Y, Jiang Y, Dehaini D, Chen M, Gu S, Gao W, Lu W, Fang RH, Zhang L. Remote-Loaded Platelet Vesicles for Disease-Targeted Delivery of Therapeutics. ADVANCED FUNCTIONAL MATERIALS 2018; 28:1801032. [PMID: 30319322 PMCID: PMC6181445 DOI: 10.1002/adfm.201801032] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Indexed: 05/18/2023]
Abstract
The recent emergence of biomimetic nanotechnology has facilitated the development of next-generation nanodelivery systems capable of enhanced biointerfacing. In particular, the direct use of natural cell membranes can enable multivalent targeting functionalities. Herein, we report on the remote loading of small molecule therapeutics into cholesterol-enriched platelet membrane-derived vesicles for disease-targeted delivery. Using this approach, high loading yields for two model drugs, doxorubicin and vancomycin, are achieved. Leveraging the surface markers found on platelet membranes, the resultant nanoformulations demonstrate natural affinity towards both breast cancer cells and methicillin-resistant Staphylococcus aureus. In vivo, this translates to improved disease targeting, increasing the potency of the encapsulated drug payloads compared with free drugs and the corresponding non-targeted nanoformulations. Overall, this work demonstrates that the remote loading of drugs into functional platelet membrane-derived vesicles is a facile means of fabricating targeted nanoformulations, an approach that can be easily generalized to other cell types in the future.
Collapse
Affiliation(s)
- Man Ying
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Jia Zhuang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Xiaoli Wei
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Xinxin Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Yue Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Yao Jiang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Diana Dehaini
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Mengchun Chen
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Silun Gu
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Weiwei Gao
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University, and Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, P.R. China
| | - Ronnie H Fang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Liangfang Zhang
- Department of NanoEngineering and Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
13
|
Neophytou C, Boutsikos P, Papageorgis P. Molecular Mechanisms and Emerging Therapeutic Targets of Triple-Negative Breast Cancer Metastasis. Front Oncol 2018. [PMID: 29520340 PMCID: PMC5827095 DOI: 10.3389/fonc.2018.00031] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Breast cancer represents a highly heterogeneous disease comprised by several subtypes with distinct histological features, underlying molecular etiology and clinical behaviors. It is widely accepted that triple-negative breast cancer (TNBC) is one of the most aggressive subtypes, often associated with poor patient outcome due to the development of metastases in secondary organs, such as the lungs, brain, and bone. The molecular complexity of the metastatic process in combination with the lack of effective targeted therapies for TNBC metastasis have fostered significant research efforts during the past few years to identify molecular “drivers” of this lethal cascade. In this review, the most current and important findings on TNBC metastasis, as well as its closely associated basal-like subtype, including metastasis-promoting or suppressor genes and aberrantly regulated signaling pathways at specific stages of the metastatic cascade are being discussed. Finally, the most promising therapeutic approaches and novel strategies emerging from these molecular targets that could potentially be clinically applied in the near future are being highlighted.
Collapse
Affiliation(s)
- Christiana Neophytou
- Department of Biological Sciences, School of Pure and Applied Sciences, University of Cyprus, Nicosia, Cyprus
| | | | | |
Collapse
|
14
|
Anisiewicz A, Pawlik A, Filip-Psurska B, Turlej E, Dzimira S, Milczarek M, Gdesz K, Papiernik D, Jarosz J, Kłopotowska D, Kutner A, Mazur A, Wietrzyk J. Unfavorable effect of calcitriol and its low-calcemic analogs on metastasis of 4T1 mouse mammary gland cancer. Int J Oncol 2017; 52:103-126. [PMID: 29115583 PMCID: PMC5743363 DOI: 10.3892/ijo.2017.4185] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 10/12/2017] [Indexed: 12/17/2022] Open
Abstract
Low vitamin D status is considered as a risk factor for breast cancer and has prognostic significance. Furthermore, vitamin D deficiency increases after adjuvant cancer therapy, which alters bone metabolism increasing the risk of osteoporosis. It is now postulated that vitamin D supplementation in breast cancer treatment delays the recurrence of cancer thereby extending survival. We evaluated the impact of calcitriol and its low-calcemic analogs, PRI-2191 and PRI-2205, on the tumor growth, angiogenesis, and metastasis of 4T1 mouse mammary gland cancer. Gene expression analysis related to cancer invasion/metastasis, real-time PCR, ELISA, western blotting, and histochemical studies were performed. In vitro studies were conducted to compare the effects of calcitriol and its analogs on 4T1 and 67NR cell proliferation and expression of selected proteins. Calcitriol and its analogs increased lung metastasis without influencing the growth of primary tumor. The levels of plasma 17β-estradiol and transforming growth factor β (TGFβ) were found to be elevated after treatment. Moreover, the results showed that tumor blood perfusion improved and osteopontin (OPN) levels increased, whereas vascular endothelial growth factor (VEGF) and TGFβ levels decreased in tumors from treated mice. All the studied treatments resulted in increased collagen content in the tumor tissue in the early step of tumor progression, and calcitriol caused an increase in collagen content in lung tissue. In addition, in vitro proliferation of 4T1 tumor cells was not found to be affected by calcitriol or its analogs in contrast to non-metastatic 67NR cells. Calcitriol and its analogs enhanced the metastatic potential of 4T1 mouse mammary gland cancer by inducing the secretion of OPN probably via host cells. In addition, OPN tumor overexpression prevailed over the decreasing tumor TGFβ level and blood vessel normalization via tumor VEGF deprivation induced by calcitriol and its analogs. Moreover, the increased plasma TGFβ and 17β-estradiol levels contributed to the facilitation of metastatic process.
Collapse
Affiliation(s)
- Artur Anisiewicz
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Agata Pawlik
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Beata Filip-Psurska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Eliza Turlej
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Stanisław Dzimira
- Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland
| | - Magdalena Milczarek
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Katarzyna Gdesz
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Diana Papiernik
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Joanna Jarosz
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Dagmara Kłopotowska
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| | - Andrzej Kutner
- Department of Pharmacology, Pharmaceutical Research Institute, 01-793 Warsaw, Poland
| | - Andrzej Mazur
- Université Clermont Auvergne, INRA, UNH, F-63000 Clermont-Ferrand, France
| | - Joanna Wietrzyk
- Department of Experimental Oncology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53‑114 Wroclaw, Poland
| |
Collapse
|
15
|
Khamrang T, Hung KC, Hsia CH, Hsieh CY, Velusamy M, Jayakumar T, Sheu JR. Antiplatelet Activity of a Newly Synthesized Novel Ruthenium (II): A Potential Role for Akt/JNK Signaling. Int J Mol Sci 2017; 18:ijms18050916. [PMID: 28448438 PMCID: PMC5454829 DOI: 10.3390/ijms18050916] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/04/2017] [Accepted: 04/18/2017] [Indexed: 01/18/2023] Open
Abstract
In oncotherapy, ruthenium complexes are considered as potential alternatives for platinum compounds, and have been proved as promising anticancer drugs with high efficacy and lesser side effects. Platelet activation plays a major role in cancer metastasis and progression. Hence, this study explored the effect of a newly synthesized ruthenium complex, [Ru(η6-cymene)(L)Cl]BF4(TQ5), where L = 4-phenyl-2-pyridin-2-yl-quinazoline), on human platelet activation. TQ5 (3–5 µM) inhibited concentration-dependent collagen-induced platelet aggregation in washed human platelets. However, this compound only inhibited platelet aggregation at a maximum concentration of 500 and 100 µM against thrombin and 9,11-dideoxy-11α, 9α-epoxymethanoprostaglandin (U46619)-induced stimulation, respectively. TQ5 inhibited collagen-induced ATP release and calcium mobilization ([Ca2+]i), without inducing cell cytotoxicity. In addition, neither SQ22536, an adenylate cyclase inhibitor, nor 1H-[1,2,4] oxadiazolo [4,3-a]quinoxalin-1-one (ODQ), a guanylate cyclase inhibitor, significantly reversed the TQ5-mediated inhibition of platelet aggregation. TQ5 inhibited the collagen-induced phosphorylation of protein kinase B (Akt) and c-Jun N-terminal kinase (JNK), but did not effectively inhibit extracellular signal-regulated kinase 1/2 (ERK1/2) and p38-mitogen-activated protein kinase (p38-MAPK) in human platelets. Additionally, TQ5 significantly prolonged the closure time in whole blood and increased the occlusion time of thrombotic platelet plug formation in mice. This study demonstrates, for the first time, that a newly synthesized ruthenium complex, TQ5, exhibits potent antiplatelet activity by hindering ATP release and [Ca2+]i, and by decreasing the activation of Akt/JNK signals. Together, these results suggest that TQ5 could be developed as a therapeutic agent that helps prevent or treat thromboembolic disorders, since it is found to be potently more effective than a well-established antithrombotic aspirin.
Collapse
Affiliation(s)
- Themmila Khamrang
- Department of Chemistry, North Eastern Hill University, Shillong 793022, India.
| | - Kuo-Chen Hung
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Gastroenterologic Surgery Division, Department of Surgery, Yuan's General Hospital, Kaohsiung 249, Taiwan.
| | - Chih-Hsuan Hsia
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Cheng-Ying Hsieh
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Marappan Velusamy
- Department of Chemistry, North Eastern Hill University, Shillong 793022, India.
| | - Thanasekaran Jayakumar
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| | - Joen-Rong Sheu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.
| |
Collapse
|
16
|
Gregório AC, Fonseca NA, Moura V, Lacerda M, Figueiredo P, Simões S, Dias S, Moreira JN. Inoculated Cell Density as a Determinant Factor of the Growth Dynamics and Metastatic Efficiency of a Breast Cancer Murine Model. PLoS One 2016; 11:e0165817. [PMID: 27820870 PMCID: PMC5098815 DOI: 10.1371/journal.pone.0165817] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/18/2016] [Indexed: 01/16/2023] Open
Abstract
4T1 metastatic breast cancer model have been widely used to study stage IV human breast cancer. However, the frequent inoculation of a large number of cells, gives rise to fast growing tumors, as well as to a surprisingly low metastatic take rate. The present work aimed at establishing the conditions enabling high metastatic take rate of the triple-negative murine 4T1 syngeneic breast cancer model. An 87% 4T1 tumor incidence was observed when as few as 500 cancer cells were implanted. 4T1 cancer cells colonized primarily the lungs with 100% efficiency, and distant lesions were also commonly identified in the mesentery and pancreas. The drastic reduction of the number of inoculated cells resulted in increased tumor doubling times and decreased specific growth rates, following a Gompertzian tumor expansion. The established conditions for the 4T1 mouse model were further validated in a therapeutic study with peguilated liposomal doxorubicin, in clinical used in the setting of metastatic breast cancer. Inoculated cell density was proven to be a key methodological aspect towards the reproducible development of macrometastases in the 4T1 mouse model and a more reliable pre-clinical assessment of antimetastatic therapies.
Collapse
Affiliation(s)
- Ana C. Gregório
- CNC—Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- IIIUC–Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Nuno A. Fonseca
- CNC—Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- FFUC—Faculty of Pharmacy, Pólo das Ciências da Saúde, University of Coimbra, Coimbra, Portugal
| | - Vera Moura
- CNC—Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- TREAT U, SA, Coimbra, Portugal
| | - Manuela Lacerda
- IPATIMUP–Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Paulo Figueiredo
- IPOFG-EPE–Portuguese Institute of Oncology Francisco Gentil, Coimbra, Portugal
| | - Sérgio Simões
- CNC—Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- FFUC—Faculty of Pharmacy, Pólo das Ciências da Saúde, University of Coimbra, Coimbra, Portugal
| | - Sérgio Dias
- IMM–Institute of Molecular Medicine, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - João Nuno Moreira
- CNC—Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- FFUC—Faculty of Pharmacy, Pólo das Ciências da Saúde, University of Coimbra, Coimbra, Portugal
- * E-mail:
| |
Collapse
|
17
|
Abstract
Extensive experimental evidence indicates that platelets contribute to tumor cell proliferation and metastasis through direct interactions and secreted bioactive proteins. Activated platelets release secretory factors that promote growth factors, chemokines, proangiogenic regulatory proteins, proteolytic enzymes and microparticles within the microenvironment to promote tumor cell growth and invasion. Furthermore, the formation of platelet-tumor cell heteroaggregates by integrin αIIbβ3 (glycoprotein IIb/IIIa) bridging plays an important role in tumor survival by forming a physical shield around tumor cells, and thereby protecting circulating tumor cells from immune-mediated lysis by natural killer (NK) cells. Tumor cells directly activate platelets by enhancing expression of surface integrins, selectins and secretion of granules, which amplify platelet aggregation. In addition to the physical coating of tumor cells, platelets release transforming growth factor-β1 (TGF-β1) that induces phenotypic changes of epithelial to mesenchymal-like transition of tumor cells, thereby facilitating their extravasation and dissemination to distant sites during metastasis. Thus, there is a complex interplay between platelet-induced tumor growth and tumor cell-induced platelet activation, with the involvement of multiple components within the tumor microenvironment that enhance metastasis. This review describes the intimate reciprocal cross-talk between platelets and tumor cells, and the various signaling pathways involved in tumor amplification, which may be potential therapeutic targets to disrupt the platelet-tumor loop to reduce metastatic processes.
Collapse
|
18
|
Kostadinova A, Topouzova-Hristova T, Momchilova A, Tzoneva R, Berger MR. Antitumor Lipids--Structure, Functions, and Medical Applications. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2015; 101:27-66. [PMID: 26572975 DOI: 10.1016/bs.apcsb.2015.08.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cell proliferation and metastasis are considered hallmarks of tumor progression. Therefore, efforts have been made to develop novel anticancer drugs that inhibit both the proliferation and the motility of tumor cells. Synthetic antitumor lipids (ATLs), which are chemically divided into two main classes, comprise (i) alkylphospholipids (APLs) and (ii) alkylphosphocholines (APCs). They represent a new entity of drugs with distinct antiproliferative properties in tumor cells. These compounds do not interfere with the DNA or mitotic spindle apparatus of the cell, instead, they incorporate into cell membranes, where they accumulate and interfere with lipid metabolism and lipid-dependent signaling pathways. Recently, it has been shown that the most commonly studied APLs inhibit proliferation by inducing apoptosis in malignant cells while leaving normal cells unaffected and are potent sensitizers of conventional chemo- and radiotherapy, as well as of electrical field therapy. APLs resist catabolic degradation to a large extent, therefore accumulate in the cell and interfere with lipid-dependent survival signaling pathways, notably PI3K-Akt and Raf-Erk1/2, and de novo phospholipid biosynthesis. They are internalized in the cell membrane via raft domains and cause downstream reactions as inhibition of cell growth and migration, cell cycle arrest, actin stress fibers collapse, and apoptosis. This review summarizes the in vitro, in vivo, and clinical trials of most common ATLs and their mode of action at molecular and biochemical levels.
Collapse
Affiliation(s)
- Aneliya Kostadinova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria.
| | | | - Albena Momchilova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Rumiana Tzoneva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria.
| | - Martin R Berger
- German Cancer Research Center, Toxicology and Chemotherapy Unit, Heidelberg, Germany
| |
Collapse
|
19
|
Nanobiotechnology for the Therapeutic Targeting of Cancer Cells in Blood. Cell Mol Bioeng 2015; 8:137-150. [PMID: 25798204 PMCID: PMC4361771 DOI: 10.1007/s12195-015-0381-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Accepted: 01/19/2015] [Indexed: 12/11/2022] Open
Abstract
During metastasis, circulating tumor cells migrate away from a primary tumor via the blood circulation to form secondary tumors in distant organs. Mounting evidence from clinical observations indicates that the number of circulating tumor cells (CTCs) in the blood correlates with the progression of solid tumors before and during chemotherapy. Beyond the well-established role of CTCs as a fluid biopsy, however, the field of targeting CTCs for the prevention or reduction of metastases has just emerged. Conventional cancer therapeutics have a relatively short circulation time in the blood which may render the killing of CTCs inefficient due to reduced exposure of CTCs to drugs. Nevertheless, over the past few decades, the development of nanoparticles and nanoformulations to improve the half-life and release profile of drugs in circulation has rejuvenated certain traditional medicines in the emerging field of CTC neutralization. This review focuses on how the principles of nanomedicine may be applied to target CTCs. Moreover, inspired by the interactions between CTCs and host cells in the blood circulation, novel biomimetic approaches for targeted drug delivery are presented.
Collapse
|
20
|
Nanobiotechnology for the Therapeutic Targeting of Cancer Cells in Blood. Cell Mol Bioeng 2015. [DOI: 10.1007/s12195-015-0378-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
21
|
Carter RZ, Micocci KC, Natoli A, Redvers RP, Paquet-Fifield S, Martin ACBM, Denoyer D, Ling X, Kim SH, Tomasin R, Selistre-de-Araújo H, Anderson RL, Pouliot N. Tumour but not stromal expression of β3 integrin is essential, and is required early, for spontaneous dissemination of bone-metastatic breast cancer. J Pathol 2015; 235:760-72. [PMID: 25430721 DOI: 10.1002/path.4490] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 11/09/2014] [Accepted: 11/25/2014] [Indexed: 02/04/2023]
Abstract
Although many preclinical studies have implicated β3 integrin receptors (αvβ3 and αIIbβ3) in cancer progression, β3 inhibitors have shown only modest efficacy in patients with advanced solid tumours. The limited efficacy of β3 inhibitors in patients could arise from our incomplete understanding of the precise function of β3 integrin and, consequently, inappropriate clinical application. Data from animal studies are conflicting and indicate heterogeneity with respect to the relative contributions of β3-expressing tumour and stromal cell populations in different cancers. Here we aimed to clarify the function and relative contributions to metastasis of tumour versus stromal β3 integrin in clinically relevant models of spontaneous breast cancer metastasis, with particular emphasis on bone metastasis. We show that stable down-regulation of tumour β3 integrin dramatically impairs spontaneous (but not experimental) metastasis to bone and lung without affecting primary tumour growth in the mammary gland. Unexpectedly, and in contrast to subcutaneous tumours, orthotopic tumour vascularity, growth and spontaneous metastasis were not altered in mice null for β3 integrin. Tumour β3 integrin promoted migration, protease expression and trans-endothelial migration in vitro and increased vascular dissemination in vivo, but was not necessary for bone colonization in experimental metastasis assays. We conclude that tumour, rather than stromal, β3 expression is essential and is required early for efficient spontaneous breast cancer metastasis to bone and soft tissues. Accordingly, differential gene expression analysis in cohorts of breast cancer patients showed a strong association between high β3 expression, early metastasis and shorter disease-free survival in patients with oestrogen receptor-negative tumours. We propose that β3 inhibitors may be more efficacious if used in a neoadjuvant setting, rather than after metastases are established.
Collapse
Affiliation(s)
- Rachel Zoe Carter
- Metastasis Research Laboratory, Peter MacCallum Cancer Centre, Melbourne, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Lim SH, Yam ML, Lam ML, Kamarulzaman FA, Samat N, Kiew LV, Chung LY, Lee HB. Photodynamic Characterization of Amino Acid Conjugated 151-Hydroxypurpurin-7-lactone for Cancer Treatment. Mol Pharm 2014; 11:3164-73. [DOI: 10.1021/mp500351s] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Siang Hui Lim
- Cancer
Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang
Jaya, Selangor, Malaysia
| | - Mun Li Yam
- Cancer
Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang
Jaya, Selangor, Malaysia
| | - May Lynn Lam
- Cancer
Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang
Jaya, Selangor, Malaysia
| | - Fadzly Azhar Kamarulzaman
- Cancer
Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang
Jaya, Selangor, Malaysia
| | - Norazwana Samat
- Cancer
Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang
Jaya, Selangor, Malaysia
| | | | | | - Hong Boon Lee
- Cancer
Research Initiatives Foundation (CARIF), Sime Darby Medical Centre, Subang
Jaya, Selangor, Malaysia
| |
Collapse
|
23
|
Bailey-Downs LC, Thorpe JE, Disch BC, Bastian A, Hauser PJ, Farasyn T, Berry WL, Hurst RE, Ihnat MA. Development and characterization of a preclinical model of breast cancer lung micrometastatic to macrometastatic progression. PLoS One 2014; 9:e98624. [PMID: 24878664 PMCID: PMC4039511 DOI: 10.1371/journal.pone.0098624] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 05/06/2014] [Indexed: 11/19/2022] Open
Abstract
Most cancer patients die with metastatic disease, thus, good models that recapitulate the natural process of metastasis including a dormancy period with micrometastatic cells would be beneficial in developing treatment strategies. Herein we report a model of natural metastasis that balances time to complete experiments with a reasonable dormancy period, which can be used to better study metastatic progression. The basis for the model is a 4T1 triple negative syngeneic breast cancer model without resection of the primary tumor. A cell titration from 500 to 15,000 GFP tagged 4T1 cells implanted into fat pad number four of immune proficient eight week female BALB/cJ mice optimized speed of the model while possessing metastatic processes including dormancy and beginning of reactivation. The frequency of primary tumors was less than 50% in animals implanted with 500–1500 cells. Although implantation with over 10,000 cells resulted in 100% primary tumor development, the tumors and macrometastases formed were highly aggressive, lacked dormancy, and offered no opportunity for treatment. Implantation of 7,500 cells resulted in >90% tumor take by 10 days; in 30–60 micrometastases in the lung (with many animals also having 2–30 brain micrometastases) two weeks post-implantation, with the first small macrometastases present at five weeks; many animals displaying macrometastases at five weeks and animals becoming moribund by six weeks post-implantation. Using the optimum of 7,500 cells the efficacy of a chemotherapeutic agent for breast cancer, doxorubicin, given at its maximal tolerated dose (MTD; 1 mg/kg weekly) was tested for an effect on metastasis. Doxorubicin treatment significantly reduced primary tumor growth and lung micrometastases but the number of macrometastases at experiment end was not significantly affected. This model should prove useful for development of drugs to target metastasis and to study the biology of metastasis.
Collapse
Affiliation(s)
| | - Jessica E. Thorpe
- DormaTarg, Inc., Oklahoma City, Oklahoma, United States of America
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, Oklahoma City, Oklahoma, United States of America
| | - Bryan C. Disch
- DormaTarg, Inc., Oklahoma City, Oklahoma, United States of America
| | - Anja Bastian
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, Oklahoma City, Oklahoma, United States of America
| | - Paul J. Hauser
- DormaTarg, Inc., Oklahoma City, Oklahoma, United States of America
- Department of Urology, University of Oklahoma College of Medicine, Oklahoma City, Oklahoma, United States of America
| | - Taleah Farasyn
- DormaTarg, Inc., Oklahoma City, Oklahoma, United States of America
| | - William L. Berry
- Department of Cell Biology, University of Oklahoma College of Medicine, Oklahoma City, Oklahoma, United States of America
| | - Robert E. Hurst
- DormaTarg, Inc., Oklahoma City, Oklahoma, United States of America
- Department of Urology, University of Oklahoma College of Medicine, Oklahoma City, Oklahoma, United States of America
- Department of Biochemistry and Molecular Biology, of Oklahoma College of Medicine, Oklahoma City, Oklahoma, United States of America
| | - Michael A. Ihnat
- DormaTarg, Inc., Oklahoma City, Oklahoma, United States of America
- Department of Pharmaceutical Sciences, University of Oklahoma College of Pharmacy, Oklahoma City, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
24
|
Ma X, Wang Y, Sheng H, Tian W, Qi Z, Teng F, Xue F. Prognostic significance of thrombocytosis, platelet parameters and aggregation rates in epithelial ovarian cancer. J Obstet Gynaecol Res 2013; 40:178-83. [PMID: 24102732 DOI: 10.1111/jog.12151] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2013] [Accepted: 04/16/2013] [Indexed: 01/12/2023]
Abstract
AIM The aim of this study is to investigate the impact of preoperative platelet counts, parameters and aggregation rates (maximal aggregation rate: MAR) on prognosis in patients with epithelial ovarian cancer (EOC). METHODS Preoperative platelet count, parameters and MAR in 182 EOC patients, 122 patients with benign ovarian tumor and 150 healthy women were retrospectively analyzed. The correlation between thrombocytosis, platelet parameters, MAR and clinicopathological factors were evaluated in EOC. RESULTS Forty-five (24.73%) EOC patients had preoperative thrombocytosis in this study. The mean platelet count in the EOC group was significantly higher than that of benign and healthy groups (P < 0.001). The MAR in the EOC group was significantly higher than that in the healthy group (71.96% vs 57.03%, P = 0.025). The platelet parameters (mean platelet volume, platelet distribution width, thrombocytocrit and large platelet ratio) were consistently higher in the EOC group than those in the benign and healthy groups, but the differences were insignificant. A significant correlation between thrombocytosis and MAR was observed in EOC patients (r = 0.694, P < 0.001). EOC patients with thrombocytosis were found to have significantly higher grade (P = 0.048), more advanced stage (P = 0.045), higher level carbohydrate antigen-125 (P = 0.007) and greater likelihood of suboptimal cytoreduction (P = 0.035). EOC patients with both thrombocytosis and high MAR were found to have shorter progression-free survival (P = 0.001)and overall survival (P = 0.004). The combination of thrombocytosis and MAR, as well as stage and optimal cytoreduction, retained significance as an independent prognostic factor for overall survival. CONCLUSION Thrombocytosis, accompanied by increasing of platelet aggregation rates, is associated with more aggressive tumor biology in EOC. The combination of thrombocytosis and MAR is an independent negative prognostic factor for overall survival in EOC patients.
Collapse
Affiliation(s)
- Xuegong Ma
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | | | | | | | | | | | | |
Collapse
|
25
|
Surgical Stress Promotes the Development of Cancer Metastases by a Coagulation-Dependent Mechanism Involving Natural Killer Cells in a Murine Model. Ann Surg 2013; 258:158-68. [DOI: 10.1097/sla.0b013e31826fcbdb] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
26
|
Levy I, Horvath A, Azevedo M, de Alexandre RB, Stratakis CA. Phosphodiesterase function and endocrine cells: links to human disease and roles in tumor development and treatment. Curr Opin Pharmacol 2011; 11:689-97. [PMID: 22047791 DOI: 10.1016/j.coph.2011.10.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Accepted: 10/04/2011] [Indexed: 12/20/2022]
Abstract
Phosphodiesterases (PDEs) are enzymes that regulate the intracellular levels of cyclic adenosine monophosphate and cyclic guanosine monophosphate, and, consequently, exhibit a central role in multiple cellular functions. The pharmacological exploitation of the ability of PDEs to regulate specific pathways has led to the discovery of drugs with selective action against specific PDE isoforms. Considerable attention has been given to the development of selective PDE inhibitors, especially after the therapeutic success of PDE5 inhibitors in the treatment of erectile dysfunction. Several associations between PDE genes and genetic diseases have been described, and more recently PDE11A and PDE8B have been implicated in predisposition to tumor formation. This review focuses on the possible function of PDEs in a variety of tumors, primarily in endocrine glands, both in tumor predisposition and as potential therapeutic targets.
Collapse
Affiliation(s)
- Isaac Levy
- Section of Endocrinology and Genetics, Program on Developmental Endocrinology Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | |
Collapse
|
27
|
Julien S, Ivetic A, Grigoriadis A, QiZe D, Burford B, Sproviero D, Picco G, Gillett C, Papp SL, Schaffer L, Tutt A, Taylor-Papadimitriou J, Pinder SE, Burchell JM. Selectin ligand sialyl-Lewis x antigen drives metastasis of hormone-dependent breast cancers. Cancer Res 2011; 71:7683-93. [PMID: 22025563 DOI: 10.1158/0008-5472.can-11-1139] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The glycome acts as an essential interface between cells and the surrounding microenvironment. However, changes in glycosylation occur in nearly all breast cancers, which can alter this interaction. Here, we report that profiles of glycosylation vary between ER-positive and ER-negative breast cancers. We found that genes involved in the synthesis of sialyl-Lewis x (sLe(x); FUT3, FUT4, and ST3GAL6) are significantly increased in estrogen receptor alpha-negative (ER-negative) tumors compared with ER-positive ones. SLe(x) expression had no influence on the survival of patients whether they had ER-negative or ER-positive tumors. However, high expression of sLe(x) in ER-positive tumors was correlated with metastasis to the bone where sLe(x) receptor E-selectin is constitutively expressed. The ER-positive ZR-75-1 and the ER-negative BT20 cell lines both express sLe(x) but only ZR-75-1 cells could adhere to activated endothelial cells under dynamic flow conditions in a sLe(x) and E-selectin-dependent manner. Moreover, L/P-selectins bound strongly to ER-negative MDA-MB-231 and BT-20 cell lines in a heparan sulfate (HS)-dependent manner that was independent of sLe(x) expression. Expression of glycosylation genes involved in heparan biosynthesis (EXT1 and HS3ST1) was increased in ER-negative tumors. Taken together, our results suggest that the context of sLe(x) expression is important in determining its functional significance and that selectins may promote metastasis in breast cancer through protein-associated sLe(x) and HS glycosaminoglycans.
Collapse
Affiliation(s)
- Sylvain Julien
- Breast Cancer Biology, King's College London, Guy's Hospital, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Traditionally viewed as major cellular components in hemostasis and thrombosis, the contribution of platelets to the progression of cancer is an emerging area of research interest. Complex interactions between tumor cells and circulating platelets play an important role in cancer growth and dissemination, and a growing body of evidence supports a role for physiologic platelet receptors and platelet agonists in cancer metastases and angiogenesis. Platelets provide a procoagulant surface facilitating amplification of cancer-related coagulation, and can be recruited to shroud tumor cells, thereby shielding them from immune responses, and facilitate cancer growth and dissemination. Experimental blockade of key platelet receptors, such as GP1b/IX/V, GPIIbIIIa and GPVI, has been shown to attenuate metastases. Platelets are also recognized as dynamic reservoirs of proangiogenic and anti-angiogenic proteins that can be manipulated pharmacologically. A bidirectional relationship between platelets and tumors is also seen, with evidence of 'tumor conditioning' of platelets. The platelet as a reporter of malignancy and a targeted delivery system for anticancer therapy has also been proposed. The development of platelet inhibitors that influence malignancy progression and clinical testing of currently available antiplatelet drugs represents a promising area of targeted cancer therapy.
Collapse
Affiliation(s)
- N M Bambace
- Division of Hematology and Oncology, Department of Medicine, University of Vermont, Burlington, VT 05401, USA
| | | |
Collapse
|
29
|
Abstract
Extensive experimental evidence shows that platelets support tumour metastasis. The activation of platelets and the coagulation system have a crucial role in the progression of cancer. Within the circulatory system, platelets guard tumour cells from immune elimination and promote their arrest at the endothelium, supporting the establishment of secondary lesions. These contributions of platelets to tumour cell survival and spread suggest platelets as a new avenue for therapy.
Collapse
Affiliation(s)
- Laurie J Gay
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|