51
|
Rittig SM, Lutz MS, Clar KL, Zhou Y, Kropp KN, Koch A, Hartkopf AD, Hinterleitner M, Zender L, Salih HR, Maurer S, Hinterleitner C. Controversial Role of the Immune Checkpoint OX40L Expression on Platelets in Breast Cancer Progression. Front Oncol 2022; 12:917834. [PMID: 35875148 PMCID: PMC9304936 DOI: 10.3389/fonc.2022.917834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/08/2022] [Indexed: 11/22/2022] Open
Abstract
In conventional T cells, OX40 has been identified as a major costimulating receptor augmenting survival and clonal expansion of effector and memory T cell populations. In regulatory T cells, (Treg) OX40 signaling suppresses cellular activity and differentiation. However, clinical trials investigating OX40 agonists to enhance anti-tumor immunity, showed only limited success so far. Here we show that platelets from breast cancer patients express relevant levels of OX40L and platelet OX40L (pOX40L) inversely correlates with platelet-expressed immune checkpoint molecules GITRL (pGITRL) and TACI (pTACI). While high expression of pOX40L correlates with T and NK cell activation, elevated pOX40L levels identify patients with higher tumor grades, the occurrence of metastases, and shorter recurrence-free survival (RFS). Of note, OX40 mRNA levels in breast cancer correlate with enhanced expression of anti-apoptotic, immune-suppressive, and tumor-promoting mRNA gene signatures. Our data suggest that OX40L on platelets might play counteracting roles in cancer and anti-tumor immunity. Since pOX40L reflects disease relapse better than the routinely used predictive markers CA15-3, CEA, and LDH, it could serve as a novel biomarker for refractory disease in breast cancer.
Collapse
Affiliation(s)
- Susanne M. Rittig
- Department of Hematology, Oncology and Cancer Immunology, Charité – Universitaetsmedizin Berlin, Corporate Member of Freie Universitaet Berlin and Humboldt-Universitaet zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité – Universitaetsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité (Junior) (Digital) Clinician Scientist Program, Berlin, Germany
| | - Martina S. Lutz
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies” , University of Tuebingen, Tuebingen, Germany
| | - Kim L. Clar
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies” , University of Tuebingen, Tuebingen, Germany
| | - Yanjun Zhou
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies” , University of Tuebingen, Tuebingen, Germany
| | - Korbinian N. Kropp
- Department of Hematology, Medical Oncology and Pneumology, University Medical Center of Mainz, Mainz, Germany
| | - André Koch
- Department of Obstetrics and Gynecology, University Hospital Tuebingen, Tuebingen, Germany
| | - Andreas D. Hartkopf
- Department of Obstetrics and Gynecology, University Hospital Tuebingen, Tuebingen, Germany
- Department of Gynecology and Obstetrics, University Hospital of Ulm, Ulm, Germany
| | - Martina Hinterleitner
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies” , University of Tuebingen, Tuebingen, Germany
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tuebingen, Tuebingen, Germany
| | - Lars Zender
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies” , University of Tuebingen, Tuebingen, Germany
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tuebingen, Tuebingen, Germany
- German Cancer Research Consortium (DKTK), Partner Site Tuebingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Helmut R. Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies” , University of Tuebingen, Tuebingen, Germany
| | - Stefanie Maurer
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies” , University of Tuebingen, Tuebingen, Germany
- Precision Immunology Institute, Department of Oncological Sciences, and The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- *Correspondence: Stefanie Maurer,
| | - Clemens Hinterleitner
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies” , University of Tuebingen, Tuebingen, Germany
- Department of Medical Oncology and Pneumology (Internal Medicine VIII), University Hospital Tuebingen, Tuebingen, Germany
- Department of Cancer Biology and Genetics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
52
|
Effects of Momordica charantia exosomes on platelet activation, adhesion, and aggregation. Blood Coagul Fibrinolysis 2022; 33:372-380. [PMID: 35834718 DOI: 10.1097/mbc.0000000000001151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The platelets play a crucial role in the progression of multiple medical conditions, such as stroke and tumor metastasis, where antiplatelet therapy may be a boon for treating these diseases. In this study, we have attempted to study the effects of extracted Momordica charantia exosomes (MCEs) on platelet activation, adhesion, and aggregation. Adult platelets isolated from healthy individuals were dose-dependently treated with MCEs (0.1, 40, and 200 μg/ml). We performed flow cytometry to detect the expression of platelet activation protein marker-activated GP IIb/IIIa (PAC-1) and P-selectin (CD62P). Platelet adhesion was analyzed through fluorescence labeling assays. The effect of MCEs on platelet-mediated cell migration of HCT116 cells was observed by transwell. Furthermore, the MCAO model of Sprague-Dawley rats was used to observe the effect of MCEs (200, 400, and 800 μg/kg) on platelet aggregation and maximum thrombotic agglutination in vivo. The results showed that 200 μg/ml MCEs exerted the most pronounced effect on platelet activation, adhesion, and aggregation. Experiments on animals showed that MCEs significantly inhibited platelet aggregation and attenuated the maximum thrombus agglutination. We concluded that MCEs inhibited platelet activation, adhesion, aggregation, and platelet-mediated migration of HCT116 cells, indicating the potential role MCEs may play in the treatment of stroke and tumor metastasis.
Collapse
|
53
|
Huang WJ, Wang GY, Liu ZY, Zhang ML, Wang W, Zhang X, Wang RT. Preoperative PDW levels predict pulmonary metastasis in patients with hepatocellular carcinoma. BMC Cancer 2022; 22:683. [PMID: 35729523 PMCID: PMC9215007 DOI: 10.1186/s12885-022-09754-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 06/10/2022] [Indexed: 12/03/2022] Open
Abstract
Background In hepatocellular carcinoma (HCC), pulmonary metastasis (PM) after hepatectomy is associated with poor clinical outcomes. The crucial phases of tumour cell proliferation, angiogenesis, and metastasis all entail platelet activation. In HCC, platelet distribution width (PDW) suggests platelet size changes and predicts a worse prognosis. The aim of this study was to assess the association between PDW and PMs in HCC patients receiving hepatectomy. Material/methods From January 2013 to December 2015, a cohort of patients who underwent hepatectomy for HCC at the Harbin Medical University Cancer Hospital in China were retrospectively evaluated. The relationship between PDW levels and clinical and demographic parameters was examined. To investigate the relationships between predicted factors and PM, a competing risk model was used. From January 2016 to December 2018, a validation cohort of 109 patients from the First Affiliated Hospital of Harbin Medical University was studied independently. Results In the primary cohort, 19 out of 214 patients had postoperative PMs. In HCC patients with PM, PDW levels were lower than in those without PM. There was a significant difference in the cumulative incidence of 2-year PM between the high-PDW and low-PDW groups after controlling for competing risk events (death prior to the development of PM) (p < 0.001). In addition, PDW was also found to be an independent predictor for PM in a multivariable competing risk analysis. The results were externally validated in another cohort. Conclusions In HCC, preoperative PDW is significantly associated with PM. PDW could be a biomarker for post-operative PM in HCC patients.
Collapse
Affiliation(s)
- Wen-Juan Huang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Guang-Yu Wang
- Department of Digestive Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Zeng-Yao Liu
- Department of Interventional Medicine, The First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, 150001, China
| | - Meng-Lin Zhang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Wen Wang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Xin Zhang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China.
| | - Rui-Tao Wang
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, Heilongjiang, 150081, China.
| |
Collapse
|
54
|
Leng Q, Ding J, Dai M, Liu L, Fang Q, Wang DW, Wu L, Wang Y. Insights Into Platelet-Derived MicroRNAs in Cardiovascular and Oncologic Diseases: Potential Predictor and Therapeutic Target. Front Cardiovasc Med 2022; 9:879351. [PMID: 35757325 PMCID: PMC9218259 DOI: 10.3389/fcvm.2022.879351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 05/24/2022] [Indexed: 11/28/2022] Open
Abstract
Non-communicable diseases (NCDs), represented by cardiovascular diseases and cancer, have been the leading cause of death globally. Improvements in mortality from cardiovascular (CV) diseases (decrease of 14%/100,000, United States) or cancers (increase 7.5%/100,000, United States) seem unsatisfactory during the past two decades, and so the search for innovative and accurate biomarkers of early diagnosis and prevention, and novel treatment strategies is a valuable clinical and economic endeavor. Both tumors and cardiovascular system are rich in angiological systems that maintain material exchange, signal transduction and distant regulation. This pattern determines that they are strongly influenced by circulating substances, such as glycolipid metabolism, inflammatory homeostasis and cyclic non-coding RNA and so forth. Platelets, a group of small anucleated cells, inherit many mature proteins, mRNAs, and non-coding RNAs from their parent megakaryocytes during gradual formation and manifest important roles in inflammation, angiogenesis, atherosclerosis, stroke, myocardial infarction, diabetes, cancer, and many other diseases apart from its classical function in hemostasis. MicroRNAs (miRNAs) are a class of non-coding RNAs containing ∼22 nucleotides that participate in many key cellular processes by pairing with mRNAs at partially complementary binding sites for post-transcriptional regulation of gene expression. Platelets contain fully functional miRNA processors in their microvesicles and are able to transport their miRNAs to neighboring cells and regulate their gene expression. Therefore, the importance of platelet-derived miRNAs for the human health is of increasing interest. Here, we will elaborate systematically the roles of platelet-derived miRNAs in cardiovascular disease and cancer in the hope of providing clinicians with new ideas for early diagnosis and therapeutic strategies.
Collapse
|
55
|
Wang X, Zhao S, Wang Z, Gao T. Platelets involved tumor cell EMT during circulation: communications and interventions. Cell Commun Signal 2022; 20:82. [PMID: 35659308 PMCID: PMC9166407 DOI: 10.1186/s12964-022-00887-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 04/24/2022] [Indexed: 12/18/2022] Open
Abstract
AbstractDistant spreading of metastatic tumor cells is still the leading cause of tumor death. Metastatic spreading is a complex process, in which epithelial-mesenchymal transition (EMT) is the primary and key event to promote it. Presently, extensive reviews have given insights on the occurrence of EMT at the primary tumor site that depends on invasive properties of tumor cells and the tumor-associated microenvironment. However, essential roles of circulation environment involved in tumor cell EMT is not well summarized. As a main constituent of the blood, platelet is increasingly found to work as an important activator to induce EMT. Therefore, this review aims to emphasize the novel role of platelet in EMT through signal communications between platelets and circulation tumor cells, and illustrate potent interventions aiming at their communications. It may give a complementary view of EMT in addition to the tissue microenvironment, help for better understand the hematogenous metastasis, and also illustrate theoretical and practical basis for the targeted inhibition.
Collapse
|
56
|
Le Chapelain O, Ho-Tin-Noé B. Intratumoral Platelets: Harmful or Incidental Bystanders of the Tumor Microenvironment? Cancers (Basel) 2022; 14:cancers14092192. [PMID: 35565321 PMCID: PMC9105443 DOI: 10.3390/cancers14092192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The tumor microenvironment (TME) is the complex and heterogenous ecosystem of solid tumors known to influence their growth and their progression. Besides tumor cells, the TME comprises a variety of host-derived cell types, ranging from endothelial cells to fibroblasts and immune cells. Clinical and experimental data are converging to indicate that platelets, originally known for their fundamental hemostatic function, also participate in tumor development and shaping of the TME. Considering the abundance of antiplatelet drugs, understanding if and how platelets contribute to the TME may lead to new therapeutic tools for improved cancer prevention and treatments. Abstract The tumor microenvironment (TME) has gained considerable interest because of its decisive impact on cancer progression, response to treatment, and disease recurrence. The TME can favor the proliferation, dissemination, and immune evasion of cancer cells. Likewise, there is accumulating evidence that intratumoral platelets could favor the development and aggressiveness of solid tumors, notably by influencing tumor cell phenotype and shaping the vascular and immune TME components. Yet, in contrast to other tumor-associated cell types like macrophages and fibroblasts, platelets are still often overlooked as components of the TME. This might be due, in part, to a deficit in investigating and reporting the presence of platelets in the TME and its relationships with cancer characteristics. This review summarizes available evidence from clinical and animal studies supporting the notion that tumor-associated platelets are not incidental bystanders but instead integral and active components of the TME. A particular emphasis is given to the description of intratumoral platelets, as well as to the functional consequences and possible mechanisms of intratumoral platelet accumulation.
Collapse
|
57
|
Zhang Y, Sun Y, Dong X, Wang QS, Zhu D, Mei L, Yan H, Lv F. A Platelet Intelligent Vehicle with Navigation for Cancer Photothermal-Chemotherapy. ACS NANO 2022; 16:6359-6371. [PMID: 35324149 DOI: 10.1021/acsnano.2c00453] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Controllable and visible delivery of therapeutic agents is critical for tumor precise therapy. Tumor targeting and deep penetration of therapeutic agents are still challenging issues for controllable delivery. Visible drug delivery with imaging navigation can optimize the treatment window for personalized medicine. Herein, a biomimetic platelet intelligent vehicle with navigation (IRDNP-PLT) was developed to achieve controllable and visible delivery with a navigation system, a driving system, and a loading system. The platelets acted as engines and drug repositories to exert the target driving and delivery functions. The fluorescent photothermal agent IR-820 was introduced in the platform to offer an imaging navigation for the intelligent platelet vehicle in addition to photothermal therapy. The nanodrug-loaded platelets enabled efficient drug loading and controlled release of the therapeutic payload by encapsulating photothermal-/pH-sensitive chemotherapeutic nanoparticles (PDA@Dox NPs). In in vivo experiments on 4T1 tumor-bearing mice models, IRDNP-PLT performed well in tumor targeting and showed excellent therapeutic efficacy and tumor recurrence prevention ability. The intelligent platelet vehicle achieved the functions of tumor targeting and deep penetration, fluorescence imaging guidance, photocontrolled drug release, and chemo-photothermal combination therapy, suggesting the advancement for tumor precise delivery and efficient therapy.
Collapse
Affiliation(s)
- Yan Zhang
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Yuanchao Sun
- Key Laboratory of Functional Polymer Materials (Ministry of Education), College of Chemistry, Nankai University, Tianjin 300071, People's Republic of China
| | - Xia Dong
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Qiang-Song Wang
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Dunwan Zhu
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Lin Mei
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Husheng Yan
- Key Laboratory of Functional Polymer Materials (Ministry of Education), College of Chemistry, Nankai University, Tianjin 300071, People's Republic of China
| | - Feng Lv
- Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| |
Collapse
|
58
|
Zhou H, Zhu L, Song J, Wang G, Li P, Li W, Luo P, Sun X, Wu J, Liu Y, Zhu S, Zhang Y. Liquid biopsy at the frontier of detection, prognosis and progression monitoring in colorectal cancer. Mol Cancer 2022; 21:86. [PMID: 35337361 PMCID: PMC8951719 DOI: 10.1186/s12943-022-01556-2] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/02/2022] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide and a leading cause of carcinogenic death. To date, surgical resection is regarded as the gold standard by the operator for clinical decisions. Because conventional tissue biopsy is invasive and only a small sample can sometimes be obtained, it is unable to represent the heterogeneity of tumor or dynamically monitor tumor progression. Therefore, there is an urgent need to find a new minimally invasive or noninvasive diagnostic strategy to detect CRC at an early stage and monitor CRC recurrence. Over the past years, a new diagnostic concept called “liquid biopsy” has gained much attention. Liquid biopsy is noninvasive, allowing repeated analysis and real-time monitoring of tumor recurrence, metastasis or therapeutic responses. With the advanced development of new molecular techniques in CRC, circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), exosomes, and tumor-educated platelet (TEP) detection have achieved interesting and inspiring results as the most prominent liquid biopsy markers. In this review, we focused on some clinical applications of CTCs, ctDNA, exosomes and TEPs and discuss promising future applications to solve unmet clinical needs in CRC patients.
Collapse
Affiliation(s)
- Hui Zhou
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China.,Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Liyong Zhu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Jun Song
- Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Guohui Wang
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Pengzhou Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Weizheng Li
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Ping Luo
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Xulong Sun
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Jin Wu
- Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Yunze Liu
- Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China
| | - Shaihong Zhu
- Department of General Surgery, Third Xiangya Hospital, Central South University, Changsha, 410013, China.
| | - Yi Zhang
- Department of General Surgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, China.
| |
Collapse
|
59
|
Li QR, Xu HZ, Xiao RC, Liu Y, Tang JM, Li J, Yu TT, Liu B, Li LG, Wang MF, Han N, Xu YH, Wang C, Komatsu N, Zhao L, Peng XC, Li TF, Chen X. Platelets are highly efficient and efficacious carriers for tumor-targeted nano-drug delivery. Drug Deliv 2022; 29:937-949. [PMID: 35319321 PMCID: PMC8956315 DOI: 10.1080/10717544.2022.2053762] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The present work aims to prove the concept of tumor-targeted drug delivery mediated by platelets. Doxorubicin (DOX) attached to nanodiamonds (ND-DOX) was investigated as the model payload drug of platelets. In vitro experiments first showed that ND-DOX could be loaded in mouse platelets in a dose-dependent manner with a markedly higher efficiency and capacity than free DOX. ND-DOX-loaded platelets (Plt@ND-DOX) maintained viability and ND-DOX could be stably held in the platelets for at least 4 hr. Next, mouse Lewis lung cancer cells were found to activate Plt@ND-DOX and thereby stimulate cargo unloading of Plt@ND-DOX. The unloaded ND-DOX was taken up by co-cultured cancer cells which consequently exhibited loss of viability, proliferation suppression and apoptosis. In vivo, Plt@ND-DOX displayed significantly prolonged blood circulation time over ND-DOX and DOX in mice, and Lewis tumor grafts demonstrated infiltration, activation and cargo unloading of Plt@ND-DOX in the tumor tissue. Consequently, Plt@ND-DOX effectively reversed the growth of Lewis tumor grafts which exhibited significant inhibition of cell proliferation and apoptosis. Importantly, Plt@ND-DOX displayed a markedly higher therapeutic potency than free DOX but without the severe systemic toxicity associated with DOX. Our findings are concrete proof of platelets as efficient and efficacious carriers for tumor-targeted nano-drug delivery with the following features: 1) large loading capacity and high loading efficiency, 2) good tolerance of cargo drug, 3) stable cargo retention and no cargo unloading in the absence of stimulation, 4) prolonged blood circulation time, and 5) excellent tumor distribution and tumor-activated drug unloading leading to high therapeutic potency and few adverse effects. Platelets hold great potential as efficient and efficacious carriers for tumor-targeted nano-drug delivery.
Collapse
Affiliation(s)
- Qi-Rui Li
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, China
| | - Hua-Zhen Xu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Rong-Cheng Xiao
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Yan Liu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Jun-Ming Tang
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Jian Li
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Ting-Ting Yu
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, China
| | - Bin Liu
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, China
| | - Liu-Gen Li
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, China
| | - Mei-Fang Wang
- Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, China
| | - Ning Han
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, China
| | - Yong-Hong Xu
- Institute of Ophthalmological Research, Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chao Wang
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Naoki Komatsu
- Graduate School of Human and Environmental Studies, Kyoto University, Kyoto, Japan
| | - Li Zhao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Xing-Chun Peng
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, China
| | - Tong-Fei Li
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China.,Hubei Key Laboratory of Embryonic Stem Cell Research, Taihe Hospital of Shiyan, Hubei University of Medicine, Shiyan, China
| | - Xiao Chen
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan, China.,Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| |
Collapse
|
60
|
Platelet Membrane: An Outstanding Factor in Cancer Metastasis. MEMBRANES 2022; 12:membranes12020182. [PMID: 35207103 PMCID: PMC8875259 DOI: 10.3390/membranes12020182] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/02/2022]
Abstract
In addition to being biological barriers where the internalization or release of biomolecules is decided, cell membranes are contact structures between the interior and exterior of the cell. Here, the processes of cell signaling mediated by receptors, ions, hormones, cytokines, enzymes, growth factors, extracellular matrix (ECM), and vesicles begin. They triggering several responses from the cell membrane that include rearranging its components according to the immediate needs of the cell, for example, in the membrane of platelets, the formation of filopodia and lamellipodia as a tissue repair response. In cancer, the cancer cells must adapt to the new tumor microenvironment (TME) and acquire capacities in the cell membrane to transform their shape, such as in the case of epithelial−mesenchymal transition (EMT) in the metastatic process. The cancer cells must also attract allies in this challenging process, such as platelets, fibroblasts associated with cancer (CAF), stromal cells, adipocytes, and the extracellular matrix itself, which limits tumor growth. The platelets are enucleated cells with fairly interesting growth factors, proangiogenic factors, cytokines, mRNA, and proteins, which support the development of a tumor microenvironment and support the metastatic process. This review will discuss the different actions that platelet membranes and cancer cell membranes carry out during their relationship in the tumor microenvironment and metastasis.
Collapse
|
61
|
Palacios-Acedo AL, Langiu M, Crescence L, Mège D, Dubois C, Panicot-Dubois L. Platelet and Cancer-Cell Interactions Modulate Cancer-Associated Thrombosis Risk in Different Cancer Types. Cancers (Basel) 2022; 14:730. [PMID: 35159000 PMCID: PMC8833365 DOI: 10.3390/cancers14030730] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 02/01/2023] Open
Abstract
The first cause of death in cancer patients, after tumoral progression itself, is thrombo-embolic disease. This cancer-associated hypercoagulability state is known as Trousseau's syndrome, and the risk for developing thrombotic events differs according to cancer type and stage, as well as within patients. Massive platelet activation by tumor cells is the key mediator of thrombus formation in Trousseau's syndrome. In this literature review, we aimed to compare the interactions between cancer cells and platelets in three different cancer types, with low, medium and high thrombotic risk. We chose oral squamous cell carcinoma for the low-thrombotic-risk, colorectal adenocarcinoma for the medium-thrombotic-risk, and pancreatic carcinoma for the high-thrombotic-risk cancer type. We showcase that understanding these interactions is of the highest importance to find new biomarkers and therapeutic targets for cancer-associated thrombosis.
Collapse
Affiliation(s)
- Ana-Luisa Palacios-Acedo
- Aix Marseille University, INSERM 1263 (Institut National de la Santé et de la Recherche), INRAE 1260 (Institut National de la Recherche Agronomique et de l’Environnement), C2VN (Center for CardioVascular and Nutrition Research), 13885 Marseille, France; (A.-L.P.-A.); (M.L.); (L.C.); (D.M.); (L.P.-D.)
| | - Mélanie Langiu
- Aix Marseille University, INSERM 1263 (Institut National de la Santé et de la Recherche), INRAE 1260 (Institut National de la Recherche Agronomique et de l’Environnement), C2VN (Center for CardioVascular and Nutrition Research), 13885 Marseille, France; (A.-L.P.-A.); (M.L.); (L.C.); (D.M.); (L.P.-D.)
| | - Lydie Crescence
- Aix Marseille University, INSERM 1263 (Institut National de la Santé et de la Recherche), INRAE 1260 (Institut National de la Recherche Agronomique et de l’Environnement), C2VN (Center for CardioVascular and Nutrition Research), 13885 Marseille, France; (A.-L.P.-A.); (M.L.); (L.C.); (D.M.); (L.P.-D.)
- Marseille University, PIVMI (Plateforme d’Imagerie Vasculaire et de Microscopie Intravitale), C2VN (Center for CardioVascular and Nutrition Research), 13385 Marseille, France
| | - Diane Mège
- Aix Marseille University, INSERM 1263 (Institut National de la Santé et de la Recherche), INRAE 1260 (Institut National de la Recherche Agronomique et de l’Environnement), C2VN (Center for CardioVascular and Nutrition Research), 13885 Marseille, France; (A.-L.P.-A.); (M.L.); (L.C.); (D.M.); (L.P.-D.)
- Department of Digestive Surgery, La Timone University Hospital, 13005 Marseille, France
| | - Christophe Dubois
- Aix Marseille University, INSERM 1263 (Institut National de la Santé et de la Recherche), INRAE 1260 (Institut National de la Recherche Agronomique et de l’Environnement), C2VN (Center for CardioVascular and Nutrition Research), 13885 Marseille, France; (A.-L.P.-A.); (M.L.); (L.C.); (D.M.); (L.P.-D.)
- Marseille University, PIVMI (Plateforme d’Imagerie Vasculaire et de Microscopie Intravitale), C2VN (Center for CardioVascular and Nutrition Research), 13385 Marseille, France
| | - Laurence Panicot-Dubois
- Aix Marseille University, INSERM 1263 (Institut National de la Santé et de la Recherche), INRAE 1260 (Institut National de la Recherche Agronomique et de l’Environnement), C2VN (Center for CardioVascular and Nutrition Research), 13885 Marseille, France; (A.-L.P.-A.); (M.L.); (L.C.); (D.M.); (L.P.-D.)
- Marseille University, PIVMI (Plateforme d’Imagerie Vasculaire et de Microscopie Intravitale), C2VN (Center for CardioVascular and Nutrition Research), 13385 Marseille, France
| |
Collapse
|
62
|
Langiu M, Palacios-Acedo AL, Crescence L, Mege D, Dubois C, Panicot-Dubois L. Neutrophils, Cancer and Thrombosis: The New Bermuda Triangle in Cancer Research. Int J Mol Sci 2022; 23:ijms23031257. [PMID: 35163180 PMCID: PMC8836160 DOI: 10.3390/ijms23031257] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/18/2022] [Accepted: 01/21/2022] [Indexed: 12/17/2022] Open
Abstract
Spontaneous venous thrombosis is often the first clinical sign of cancer, and it is linked to a worsened survival rate. Traditionally, tumor-cell induced platelet activation has been the main actor studied in cancer-associated-thrombosis. However, platelet involvement alone does not seem to be sufficient to explain this heightened pro-thrombotic state. Neutrophils are emerging as key players in both thrombus generation and cancer progression. Neutrophils can impact thrombosis through the release of pro-inflammatory cytokines and expression of molecules like P-selectin and Tissue Factor (TF) on their membrane and on neutrophil-derived microvesicles. Their role in cancer progression is evidenced by the fact that patients with high blood-neutrophil counts have a worsened prognosis. Tumors can attract neutrophils to the cancer site via pro-inflammatory cytokine secretions and induce a switch to pro-tumoral (or N2) neutrophils, which support metastatic spread and have an immunosuppressive role. They can also expel their nuclear contents to entrap pathogens forming Neutrophil Extracellular Traps (NETs) and can also capture coagulation factors, enhancing the thrombus formation. These NETs are also known to have pro-tumoral effects by supporting the metastatic process. Here, we strived to do a comprehensive literature review of the role of neutrophils as drivers of both cancer-associated thrombosis (CAT) and cancer progression.
Collapse
Affiliation(s)
- Mélanie Langiu
- Aix Marseille Univ INSERM, INRAE, C2VN, 13005 Marseille, France; (M.L.); (A.-L.P.-A.); (L.C.); (D.M.); (L.P.-D.)
| | - Ana-Luisa Palacios-Acedo
- Aix Marseille Univ INSERM, INRAE, C2VN, 13005 Marseille, France; (M.L.); (A.-L.P.-A.); (L.C.); (D.M.); (L.P.-D.)
| | - Lydie Crescence
- Aix Marseille Univ INSERM, INRAE, C2VN, 13005 Marseille, France; (M.L.); (A.-L.P.-A.); (L.C.); (D.M.); (L.P.-D.)
| | - Diane Mege
- Aix Marseille Univ INSERM, INRAE, C2VN, 13005 Marseille, France; (M.L.); (A.-L.P.-A.); (L.C.); (D.M.); (L.P.-D.)
- Department of Digestive Surgery, La Timone University Hospital, 13005 Marseille, France
| | - Christophe Dubois
- Aix Marseille Univ INSERM, INRAE, C2VN, 13005 Marseille, France; (M.L.); (A.-L.P.-A.); (L.C.); (D.M.); (L.P.-D.)
- Correspondence:
| | - Laurence Panicot-Dubois
- Aix Marseille Univ INSERM, INRAE, C2VN, 13005 Marseille, France; (M.L.); (A.-L.P.-A.); (L.C.); (D.M.); (L.P.-D.)
| |
Collapse
|
63
|
Coagulome and the tumor microenvironment: an actionable interplay. Trends Cancer 2022; 8:369-383. [PMID: 35027336 DOI: 10.1016/j.trecan.2021.12.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/19/2021] [Accepted: 12/15/2021] [Indexed: 12/14/2022]
Abstract
Human tumors often trigger a hypercoagulable state that promotes hemostatic complications, including venous thromboembolism. The recent application of systems biology to the study of the coagulome highlighted its link to shaping the tumor microenvironment (TME), both within and outside of the vascular space. Addressing this link provides the opportunity to revisit the significance of biomarkers of hemostasis and assess the communication between vasculature and tumor parenchyma, an important topic considering the advent of immune checkpoint inhibitors and vascular normalization strategies. Understanding how the coagulome and TME influence each other offers exciting new prospects for predicting hemostatic complications and boosting the effectiveness of cancer treatment.
Collapse
|
64
|
Martins Castanheira N, Spanhofer AK, Wiener S, Bobe S, Schillers H. Uptake of platelets by cancer cells and recycling of the platelet protein CD42a. J Thromb Haemost 2022; 20:170-181. [PMID: 34592045 DOI: 10.1111/jth.15543] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND It is well accepted that the bidirectional crosstalk between platelets and cancer cells promotes tumorigenesis and metastasis. In an early step, cancer cells trigger platelet granule and extracellular vesicle release that is needed to facilitate cancer cell survival in circulation. OBJECTIVES To discover the early crosstalk of cancer cells and platelets. METHODS Cancer cells were incubated with freshly isolated and stained human platelets. Confocal laser scanning microscopy and flow cytometry was used to visualize and to quantify platelet uptake and the membrane presence of CD42 on cancer cells. Dyngo4a was used to test if platelet uptake is a dynamin-dependent process. RESULTS We found a dynamin-dependent uptake of platelets by cancer cells. This is followed by the recycling of the platelet-specific protein CD42a and its incorporation into cancer cells' plasma membrane, which is not a result of platelet RNA transfer by platelet-derived microparticles and exosomes. Time course of platelet uptake follows a sigmoid function revealing that 50% of the cancer cells are positive for platelets after approximately 38 min. Platelet uptake was observed for the tested cancerous cells (A549, MCF-7, and MV3) but not for the non-cancerous cell line 16HBE14o-. CONCLUSIONS Our results demonstrate that cancer cells hijack platelets by phagocytosis and recycling of platelet membrane proteins. The uptake of platelets has additional advantages for cancer cells: access to the entire and undiluted platelet proteome, transcriptome, and secretome. These novel findings will allow further mechanistic elucidation and thus help us gain deeper insights into platelet-assisted hematogenous metastasis.
Collapse
Affiliation(s)
| | - Anna K Spanhofer
- Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Sebastian Wiener
- Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Stefanie Bobe
- Institute of Physiology II, University of Muenster, Muenster, Germany
- Gerhard-Domagk-Institute of Pathology, University Hospital Muenster, Muenster, Germany
| | - Hermann Schillers
- Institute of Physiology II, University of Muenster, Muenster, Germany
| |
Collapse
|
65
|
Faria AVS, Yu B, Mommersteeg M, de Souza-Oliveira PF, Andrade SS, Spaander MCW, de Maat MPM, Peppelenbosch MP, Ferreira-Halder CV, Fuhler GM. Platelet-dependent signaling and Low Molecular Weight Protein Tyrosine Phosphatase expression promote aggressive phenotypic changes in gastrointestinal cancer cells. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166280. [PMID: 34610471 DOI: 10.1016/j.bbadis.2021.166280] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 12/12/2022]
Abstract
Over the last decades, some members of the protein tyrosine phosphatase family have emerged as cancer promoters. Among them, the Low Molecular Weight Protein Tyrosine Phosphatase (LMWPTP) has been described to be associated with colorectal cancer liver metastasis and poor prostate cancer prognosis. Of importance in the process of cancer progression and metastasis is the interaction between tumor cells and platelets, as the latter are thought to promote several tumor hallmarks. Here, we examine to what extent LMWPTP expression in tumor cells affects their interaction with platelets. We demonstrate that the gene encoding LMWPTP is overexpressed in upper gastrointestinal (GI) cancer cell as well as colorectal cancer, and subsequently employ cell line models to show that the level of this phosphatase may be further augmented in the presence of platelets. We demonstrate that tumor-platelet interaction promotes GI tumor cell proliferation. Additionally, using know-down/-out models we show that LMWPTP expression in cancer cells contributes to a more efficient interaction with platelets and drives platelet-induced proliferation. These data are the first to demonstrate that phosphatases play a positive role in the tumor-promoting activities of platelets, with LMWPTP emerging as a key player promoting oncogenic phenotypic changes in tumor cells.
Collapse
Affiliation(s)
- Alessandra V S Faria
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA Rotterdam, the Netherlands; Department of Biochemistry and Tissue Biology, University of Campinas, UNICAMP, Campinas, SP 13083-862, Brazil
| | - Bingting Yu
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA Rotterdam, the Netherlands
| | - Michiel Mommersteeg
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA Rotterdam, the Netherlands
| | | | | | - Manon C W Spaander
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA Rotterdam, the Netherlands
| | - Moniek P M de Maat
- Department of Hematology, Erasmus University Medical Center Rotterdam, NL-3000 CA Rotterdam, the Netherlands
| | - Maikel P Peppelenbosch
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA Rotterdam, the Netherlands
| | - Carmen V Ferreira-Halder
- Department of Biochemistry and Tissue Biology, University of Campinas, UNICAMP, Campinas, SP 13083-862, Brazil.
| | - Gwenny M Fuhler
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center Rotterdam, NL-3000 CA Rotterdam, the Netherlands.
| |
Collapse
|
66
|
Biomimetic platelet membrane-coated Nanoparticles for targeted therapy. Eur J Pharm Biopharm 2022; 172:1-15. [DOI: 10.1016/j.ejpb.2022.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 12/18/2021] [Accepted: 01/17/2022] [Indexed: 02/08/2023]
|
67
|
Zhang L, Bu P. The two sides of creatine in cancer. Trends Cell Biol 2021; 32:380-390. [PMID: 34895811 DOI: 10.1016/j.tcb.2021.11.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 12/23/2022]
Abstract
Creatine is a nitrogen-containing organic acid naturally existing in mammals. It can be converted into phosphocreatine to provide energy for muscle and nerve tissues. Creatine and its analog, cyclocreatine, have been considered cancer suppressive metabolites due to their effects on suppression of subcutaneous cancer growth. Recently, emerging studies have demonstrated the promoting effect of creatine on cancer metastasis. Orthotopic mouse models revealed that creatine promoted invasion and metastasis of pancreatic cancer, colorectal cancer, and breast cancer. Thus, creatine possesses considerably complicated roles in cancer progression. In this review, we systematically summarized the role of creatine in tumor progression, which will call to caution when considering creatine supplementation to clinically treat cancer patients.
Collapse
Affiliation(s)
- Liwen Zhang
- Key Laboratory of RNA Biology, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Pengcheng Bu
- Key Laboratory of RNA Biology, Key Laboratory of Protein and Peptide Pharmaceutical, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
68
|
Mai S, Inkielewicz-Stepniak I. Pancreatic Cancer and Platelets Crosstalk: A Potential Biomarker and Target. Front Cell Dev Biol 2021; 9:749689. [PMID: 34858977 PMCID: PMC8631477 DOI: 10.3389/fcell.2021.749689] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022] Open
Abstract
Platelets have been recognized as key players in hemostasis, thrombosis, and cancer. Preclinical and clinical researches evidenced that tumorigenesis and metastasis can be promoted by platelets through a wide variety of crosstalk between cancer cells and platelets. Pancreatic cancer is a devastating disease with high morbidity and mortality worldwide. Although the relationship between pancreatic cancer and platelets in clinical diagnosis is described, the interplay between pancreatic cancer and platelets, the underlying pathological mechanism and pathways remain a matter of intensive study. This review summaries recent researches in connections between platelets and pancreatic cancer. The existing data showed different underlying mechanisms were involved in their complex crosstalk. Typically, pancreatic tumor accelerates platelet aggregation which forms thrombosis. Furthermore, extracellular vesicles released by platelets promote communication in a neoplastic microenvironment and illustrate how these interactions drive disease progression. We also discuss the advantages of novel model organoids in pancreatic cancer research. A more in-depth understanding of tumor and platelets crosstalk which is based on organoids and translational therapies may provide potential diagnostic and therapeutic strategies for pancreatic cancer progression.
Collapse
Affiliation(s)
- Shaoshan Mai
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| | - Iwona Inkielewicz-Stepniak
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
69
|
Tumour microenvironment: a non-negligible driver for epithelial-mesenchymal transition in colorectal cancer. Expert Rev Mol Med 2021; 23:e16. [PMID: 34758892 DOI: 10.1017/erm.2021.13] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer remains the leading cause of death worldwide, and metastasis is still the major cause of treatment failure for cancer patients. Epithelial-mesenchymal transition (EMT) has been shown to play a critical role in the metastasis cascade of epithelium-derived carcinoma. Tumour microenvironment (TME) refers to the local tissue environment in which tumour cells produce and live, including not only tumour cells themselves, but also fibroblasts, immune and inflammatory cells, glial cells and other cells around them, as well as intercellular stroma, micro vessels and infiltrated biomolecules from the nearby areas, which has been proved to widely participate in the occurrence and progress of cancer. Emerging and accumulating studies indicate that, on one hand, mesenchymal cells in TME can establish 'crosstalk' with tumour cells to regulate their EMT programme; on the other, EMT-tumour cells can create a favourable environment for their own growth via educating stromal cells. Recently, our group has conducted a series of studies on the interaction between tumour-associated macrophages (TAMs) and colorectal cancer (CRC) cells in TME, confirming that the interaction between TAMs and CRC cells mediated by cytokines or exosomes can jointly promote the metastasis of CRC by regulating the EMT process of tumour cells and the M2-type polarisation process of TAMs. Herein, we present an overview to describe the current knowledge about EMT in cancer, summarise the important role of TME in EMT, and provide an update on the mechanisms of TME-induced EMT in CRC, aiming to provide new ideas for understanding and resisting tumour metastasis.
Collapse
|
70
|
Circulating Tumour Cells (CTCs) in NSCLC: From Prognosis to Therapy Design. Pharmaceutics 2021; 13:pharmaceutics13111879. [PMID: 34834295 PMCID: PMC8619417 DOI: 10.3390/pharmaceutics13111879] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/27/2021] [Accepted: 10/30/2021] [Indexed: 02/08/2023] Open
Abstract
Designing optimal (neo)adjuvant therapy is a crucial aspect of the treatment of non-small-cell lung carcinoma (NSCLC). Standard methods of chemotherapy, radiotherapy, and immunotherapy represent effective strategies for treatment. However, in some cases with high metastatic activity and high levels of circulating tumour cells (CTCs), the efficacy of standard treatment methods is insufficient and results in treatment failure and reduced patient survival. CTCs are seen not only as an isolated phenomenon but also a key inherent part of the formation of metastasis and a key factor in cancer death. This review discusses the impact of NSCLC therapy strategies based on a meta-analysis of clinical studies. In addition, possible therapeutic strategies for repression when standard methods fail, such as the administration of low-toxicity natural anticancer agents targeting these phenomena (curcumin and flavonoids), are also discussed. These strategies are presented in the context of key mechanisms of tumour biology with a strong influence on CTC spread and metastasis (mechanisms related to tumour-associated and -infiltrating cells, epithelial–mesenchymal transition, and migration of cancer cells).
Collapse
|
71
|
Palacios-Acedo AL, Mezouar S, Mège D, Crescence L, Dubois C, Panicot-Dubois L. P2RY12-Inhibitors Reduce Cancer-Associated Thrombosis and Tumor Growth in Pancreatic Cancers. Front Oncol 2021; 11:704945. [PMID: 34589424 PMCID: PMC8475274 DOI: 10.3389/fonc.2021.704945] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 08/19/2021] [Indexed: 12/21/2022] Open
Abstract
Platelet function can be modified by cancer cells to support tumor growth, causing alterations in the delicate hemostatic equilibrium. Cancer-cell and platelet interactions are one of the main pillars of Trousseau’s syndrome: a paraneoplastic syndrome with recurring and migrating episodes of thrombophlebitis. Altogether, this leads to a four-fold risk of thrombotic events in cancer patients, which in turn, portend a poor prognosis. We previously demonstrated that anti-P2RY12 drugs inhibit cancer-associated-thrombosis and formation of tumor metastasis in pancreatic cancer models. Here, we aimed to (1) compare the effects of aspirin and clopidogrel on pancreatic cancer prevention, (2) characterize the effects of clopidogrel (platelet P2RY12 inhibitor) on cancer-associated thrombosis and cancer growth in vivo, (3) determine the effect of P2RY12 across different digestive-tract cancers in vitro, and (4) analyze the expression pattern of P2RY12 in two different cancer types affecting the digestive system. Clopidogrel treatment resulted in better survival rates with smaller primary tumors and less metastasis than aspirin treatment. Clopidogrel was also more effective than aspirin at dissolving spontaneous endogenous thrombi in our orthotopic advanced cancer mouse model. P2RY12 expression gives pancreatic adenocarcinomas proliferative advantages. In conclusion, we propose the hypothesis that clopidogrel should be further studied to target and prevent Trousseau’s syndrome; as well as diminish cancer growth and spread. However, more studies are required to determine the implicated pathways and effects of these drugs on cancer development.
Collapse
Affiliation(s)
- Ana Luisa Palacios-Acedo
- Aix Marseille Univ, INSERM 1263, INRA 1260, Center for Cardiovascular and Nutrition Research (C2VN), Marseille, France
| | - Soraya Mezouar
- Aix Marseille Univ, INSERM 1263, INRA 1260, Center for Cardiovascular and Nutrition Research (C2VN), Marseille, France
| | - Diane Mège
- Aix Marseille Univ, INSERM 1263, INRA 1260, Center for Cardiovascular and Nutrition Research (C2VN), Marseille, France.,Department of Digestive Surgery, Timone University Hospital, Marseille, France
| | - Lydie Crescence
- Aix Marseille Univ, INSERM 1263, INRA 1260, Center for Cardiovascular and Nutrition Research (C2VN), Marseille, France
| | - Christophe Dubois
- Aix Marseille Univ, INSERM 1263, INRA 1260, Center for Cardiovascular and Nutrition Research (C2VN), Marseille, France
| | - Laurence Panicot-Dubois
- Aix Marseille Univ, INSERM 1263, INRA 1260, Center for Cardiovascular and Nutrition Research (C2VN), Marseille, France
| |
Collapse
|
72
|
Miao Y, Xu Z, Feng W, Zheng M, Xu Z, Gao H, Li W, Zhang Y, Zong Y, Lu A, Zhao J. Platelet infiltration predicts survival in postsurgical colorectal cancer patients. Int J Cancer 2021; 150:509-520. [PMID: 34551136 DOI: 10.1002/ijc.33816] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 01/23/2023]
Abstract
Platelets promote tumor growth and metastasis in several tumor types. Recent research has found platelets can extravasate and infiltrate into the tumor stroma and interact with the tumor microenvironment. The prognostic role of platelet infiltration in colorectal cancer (CRC) remains controversial. A pan-cancer survival analysis was performed to find the potential prognostic value of platelet infiltration in patients with cancer. A survival analysis and a nomogram prognostic model were established to further confirm the results with data from our center. The correlations between patient outcomes and tumor-infiltrating platelets (TIPs) were identified by immunohistochemical staining for CD42b. The prognostic accuracy and discriminative ability of the nomogram were determined by the concordance index (C-index) and a calibration curve. The pan-cancer survival analysis showed platelet infiltration can lead to a poor prognosis in patients with several types of cancers, including CRC. Platelet infiltration was associated with overall survival (OS) and disease-free survival (DFS) in both primary and validation cohorts. The C-index values of the nomogram for predicting OS and DFS were 0.774 and 0.769, respectively, which were higher than that of the TNM staging system alone. Our study found platelet infiltration has a potential prognostic value regarding postsurgical survival in CRC patients. The proposed nomogram resulted in a more accurate prognostic prediction for postsurgical CRC patients.
Collapse
Affiliation(s)
- Yiming Miao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zifeng Xu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wenqing Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Minhua Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhuoqing Xu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Han Gao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wenchang Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yuchen Zhang
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yaping Zong
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Aiguo Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jingkun Zhao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Minimally Invasive Surgery Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
73
|
Wu W, Wang Q, Han D, Li J, Nie Y, Guo D, Yang L, Tao K, Zhang X, Dou K. Prognostic value of preoperative inflammatory markers in patients with hepatocellular carcinoma who underwent curative resection. Cancer Cell Int 2021; 21:500. [PMID: 34535132 PMCID: PMC8447627 DOI: 10.1186/s12935-021-02204-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 09/07/2021] [Indexed: 02/08/2023] Open
Abstract
Background The prognosis of hepatocellular carcinoma (HCC) is not optimistic. Our study focused on present inflammatory markers, including the neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), gamma-glutamyl transpeptidase-to-platelet ratio (GPR), aspartate aminotransferase-to-lymphocyte ratio (ALR) and fibrinogen-to-albumin ratio (FAR), and explored their optimal combination for the prognosis of HCC after resection. Methods A total of 347 HCC patients who underwent curative resection were enrolled. The optimal cutoff values of the inflammatory markers were calculated using receiver operating characteristic (ROC) curve analysis, and used to divide patients into two groups whose differences were compared by Kaplan–Meier analysis. Cox univariate and multivariate analyses were used to analyze the independent prognostic inflammatory markers. The χ2 test was chosen to determine the relationship between independent prognostic inflammatory markers and clinicopathological features. We created combined scoring models and evaluated them by Cox univariate and multivariate methods. The concordance index (C-index), Akaike information criterion (AIC) and likelihood ratio were calculated to compare the models. The selected optimal inflammatory markers and their combinations were tested in different stages of HCC by Kaplan–Meier analysis. Results The ALR and GPR were independent prognostic factors for disease-free survival (DFS); the ALR, PLR, and GPR were independent prognostic factors for overall survival (OS). The proposed GPR and ALR-GPR-PLR score models were independent predictors for DFS and OS, respectively. Conclusion The preoperative GPR and ALR-GPR-PLR score models were independent predictors for DFS and OS, respectively, and performed well in stratifying patients with HCC. The higher the score in the model was, the worse the prognosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02204-3.
Collapse
Affiliation(s)
- Wenlong Wu
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Quancheng Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Dandan Han
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Jianhui Li
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Ye Nie
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Dongnan Guo
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Long Yang
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Kaishan Tao
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| | - Xuan Zhang
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| | - Kefeng Dou
- Department of Hepatobiliary Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
74
|
Matveeva K, Zyubin A, Demishkevich E, Rafalskiy V, Moiseeva E, Kon I, Kundalevich A, Butova V, Samusev I. Spectral and time-resolved photoluminescence of human platelets doped with platinum nanoparticles. PLoS One 2021; 16:e0256621. [PMID: 34469464 PMCID: PMC8409683 DOI: 10.1371/journal.pone.0256621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/10/2021] [Indexed: 11/19/2022] Open
Abstract
This paper describes a detailed study of spectral and time-resolved photoprocesses in human platelets and their complexes with platinum (Pt) nanoparticles (NPs). Fluorescence, quantum yield, and platelet amino acid lifetime changes in the presence and without femtosecond ablated platinum NPs have been studied. Fluorescence spectroscopy analysis of main fluorescent amino acids and their residues (tyrosine (Tyr), tryptophan (Trp), and phenylalanine (Phe)) belonging to the platelet membrane have been performed. The possibility of energy transfer between Pt NPs and the platelet membrane has been revealed. Förster Resonance Energy Transfer (FRET) model was used to perform the quantitative evaluation of energy transfer parameters. The prospects of Pt NPs usage deals with quenching-based sensing for pathology's based on platelet conformations as cardiovascular diseases have been demonstrated.
Collapse
Affiliation(s)
- Karina Matveeva
- REC «Fundamental and Applied Photonics, Nanophotonics», Immanuel Kant Baltic Federal University, Kaliningrad, Kaliningrad region, Russia
| | - Andrey Zyubin
- REC «Fundamental and Applied Photonics, Nanophotonics», Immanuel Kant Baltic Federal University, Kaliningrad, Kaliningrad region, Russia
| | - Elizaveta Demishkevich
- REC «Fundamental and Applied Photonics, Nanophotonics», Immanuel Kant Baltic Federal University, Kaliningrad, Kaliningrad region, Russia
| | - Vladimir Rafalskiy
- REC «Fundamental and Applied Photonics, Nanophotonics», Immanuel Kant Baltic Federal University, Kaliningrad, Kaliningrad region, Russia
| | - Ekaterina Moiseeva
- REC «Fundamental and Applied Photonics, Nanophotonics», Immanuel Kant Baltic Federal University, Kaliningrad, Kaliningrad region, Russia
| | - Igor Kon
- REC «Fundamental and Applied Photonics, Nanophotonics», Immanuel Kant Baltic Federal University, Kaliningrad, Kaliningrad region, Russia
| | - Anna Kundalevich
- REC «Fundamental and Applied Photonics, Nanophotonics», Immanuel Kant Baltic Federal University, Kaliningrad, Kaliningrad region, Russia
| | - Viktoria Butova
- REC «Fundamental and Applied Photonics, Nanophotonics», Immanuel Kant Baltic Federal University, Kaliningrad, Kaliningrad region, Russia
| | - Ilia Samusev
- REC «Fundamental and Applied Photonics, Nanophotonics», Immanuel Kant Baltic Federal University, Kaliningrad, Kaliningrad region, Russia
| |
Collapse
|
75
|
Abstract
Drug delivery systems have shown tremendous promise to improve the diagnostic and therapeutic effects of drugs due to their special property. Targeting tissue damage, tumors, or drugs with limited toxicity at the site of infection is the goal of successful pharmaceuticals. Targeted drug delivery has become significantly important in enhancing the pharmaceutical effects of drugs and reducing their side effects of therapeutics in the treatment of various disease conditions. Unfortunately, clinical translation of these targeted drug delivery system mechanisms faces many challenges. At present, only a few targeted drug delivery systems can achieve high targeting efficiency after intravenous injection, even though numerous surface markers and targeting approaches have been developed. Thus, cell-mediated drug-delivery targeting systems have received considerable attention for their enhanced therapeutic specificity and efficacy in the treatment of the disease. This review highlights the recent advances in the design of the different types of cells that have been explored for cell-mediated drug delivery and targeting mechanisms. A better understanding of cell biology orientation and a new generation of delivery strategies that utilize these endogenous approaches are expected to provide better solutions for specific site delivery and further facilitate clinical translation.
Collapse
Affiliation(s)
- Hongli Yu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Zhihong Yang
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| | - Fei Li
- Department of Pharmacy, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lisa Xu
- School of Public Health, Qingdao University, Qingdao, China
| | - Yong Sun
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
| |
Collapse
|
76
|
Jin J, Lin J, Xu A, Lou J, Qian C, Li X, Wang Y, Yu W, Tao H. CCL2: An Important Mediator Between Tumor Cells and Host Cells in Tumor Microenvironment. Front Oncol 2021; 11:722916. [PMID: 34386431 PMCID: PMC8354025 DOI: 10.3389/fonc.2021.722916] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor microenvironment (TME) formation is a major cause of immunosuppression. The TME consists of a considerable number of macrophages and stromal cells that have been identified in multiple tumor types. CCL2 is the strongest chemoattractant involved in macrophage recruitment and a powerful initiator of inflammation. Evidence indicates that CCL2 can attract other host cells in the TME and direct their differentiation in cooperation with other cytokines. Overall, CCL2 has an unfavorable effect on prognosis in tumor patients because of the accumulation of immunosuppressive cell subtypes. However, there is also evidence demonstrating that CCL2 enhances the anti-tumor capability of specific cell types such as inflammatory monocytes and neutrophils. The inflammation state of the tumor seems to have a bi-lateral role in tumor progression. Here, we review works focusing on the interactions between cancer cells and host cells, and on the biological role of CCL2 in these processes.
Collapse
Affiliation(s)
- Jiakang Jin
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Jinti Lin
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Ankai Xu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Jianan Lou
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Chao Qian
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Xiumao Li
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Yitian Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wei Yu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Huimin Tao
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
77
|
Ge X, Yuan L, Cheng B, Dai K. Identification of seven tumor-educated platelets RNAs for cancer diagnosis. J Clin Lab Anal 2021; 35:e23791. [PMID: 33955587 PMCID: PMC8183939 DOI: 10.1002/jcla.23791] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/25/2021] [Accepted: 04/01/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Tumor-educated platelets (TEPs) may enable blood-based cancer diagnosis. This study aimed to identify diagnostic TEPs genes involved in carcinogenesis. MATERIALS AND METHODS The TEPs differentially expressed genes (DEGs) between healthy samples and early/advanced cancer samples were obtained using bioinformatics. Gene ontology (GO) analysis and Kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analysis were used to identify the pathways and functional annotation of TEPs DEGs. Protein-protein interaction of these TEPs DEGs was analyzed based on the STRING database and visualized by Cytoscape software. The correlation analysis and diagnostic analysis were performed to evaluate the diagnostic value of TEPs mRNAs expression for early/advanced cancers. Quantitative real-time PCR (qRT-PCR) was applied to validate the role of DEGs in cancers. RESULTS TEPs mRNAs were mostly involved in protein binding, extracellular matrix, and cellular protein metabolic process. RSL24D1 was negatively correlated to early-stage cancers compared to healthy controls and may be potentially used for early cancer diagnosis. In addition, HPSE, IFI27, LGALS3BP, CRYM, HBD, COL6A3, LAMB2, and IFITM3 showed an upward trend in the expression from early to advanced cancer stages. Moreover, ARL2, FCGR2A, and KLHDC8B were positively associated with advanced, metastatic cancers compared to healthy controls. Among the 12 selected DEGs, the expression of 7 DEGs, including RSL24D1, IFI27, CRYM, HBD, IFITM3, FCGR2A, and KLHDC8B, were verified by the qRT-PCR method. CONCLUSION This study suggests that the 7-gene TEPs liquid-biopsy biomarkers may be used for cancer diagnosis and monitoring.
Collapse
Affiliation(s)
- Xinxin Ge
- The First Affiliated Hospital and Collaborative Innovation Center of HematologyJiangsu Institute of HematologyCyrus Tang Medical InstituteState Key Laboratory of Radiation Medicine and ProtectionKey Laboratory of Thrombosis and HemostasisMinistry of HealthNational Clinical Research Center for Hematological DiseasesSoochow UniversitySuzhouChina
| | - Liuxia Yuan
- The First Affiliated Hospital and Collaborative Innovation Center of HematologyJiangsu Institute of HematologyCyrus Tang Medical InstituteState Key Laboratory of Radiation Medicine and ProtectionKey Laboratory of Thrombosis and HemostasisMinistry of HealthNational Clinical Research Center for Hematological DiseasesSoochow UniversitySuzhouChina
| | - Bin Cheng
- The First Affiliated Hospital and Collaborative Innovation Center of HematologyJiangsu Institute of HematologyCyrus Tang Medical InstituteState Key Laboratory of Radiation Medicine and ProtectionKey Laboratory of Thrombosis and HemostasisMinistry of HealthNational Clinical Research Center for Hematological DiseasesSoochow UniversitySuzhouChina
| | - Kesheng Dai
- The First Affiliated Hospital and Collaborative Innovation Center of HematologyJiangsu Institute of HematologyCyrus Tang Medical InstituteState Key Laboratory of Radiation Medicine and ProtectionKey Laboratory of Thrombosis and HemostasisMinistry of HealthNational Clinical Research Center for Hematological DiseasesSoochow UniversitySuzhouChina
| |
Collapse
|
78
|
Dymicka-Piekarska V, Koper-Lenkiewicz OM, Zińczuk J, Kratz E, Kamińska J. Inflammatory cell-associated tumors. Not only macrophages (TAMs), fibroblasts (TAFs) and neutrophils (TANs) can infiltrate the tumor microenvironment. The unique role of tumor associated platelets (TAPs). Cancer Immunol Immunother 2021; 70:1497-1510. [PMID: 33146401 PMCID: PMC8139882 DOI: 10.1007/s00262-020-02758-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 10/15/2020] [Indexed: 12/13/2022]
Abstract
It is well known that various inflammatory cells infiltrate cancer cells. Next to TAMs (tumor-associated macrophages), TAFs (tumor-associated fibroblasts) and TANs (tumor-associated neutrophils) also platelets form the tumor microenvironment. Taking into account the role of platelets in the development of cancer, we have decided to introduce a new term: tumor associated platelets-TAPs. To the best of our knowledge, thus far this terminology has not been employed by anyone. Platelets are the first to appear at the site of the inflammatory process that accompanies cancer development. Within the first few hours from the start of the colonization of cancer cells platelet-tumor aggregates are responsible for neutrophils recruitment, and further release a number of factors associated with tumor growth, metastasis and neoangiogenesis. On the other hand, it also has been indicated that factors delivered from platelets can induce a cytotoxic effect on the proliferating neoplastic cells, and even enhance apoptosis. Undoubtedly, TAPs' role seems to be more complex when compared to tumor associated neutrophils and macrophages, which do not allow for their division into TAP P1 and TAP P2, as in the case of TANs and TAMs. In this review we discuss the role of TAPs as an important element of tumor invasiveness and as a potentially new therapeutic target to prevent cancer development. Nevertheless, better exploring the interactions between platelets and tumor cells could help in the formulation of new therapeutic goals that support or improve the effectiveness of cancer treatment.
Collapse
Affiliation(s)
- Violetta Dymicka-Piekarska
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Waszyngtona 15A, 15-269 Bialystok, Poland
| | - Olga M. Koper-Lenkiewicz
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Waszyngtona 15A, 15-269 Bialystok, Poland
| | - Justyna Zińczuk
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Waszyngtona 15A, 15-269 Bialystok, Poland
| | - Ewa Kratz
- Department of Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, Borowska Street 211A, 50-556 Wrocław, Poland
| | - Joanna Kamińska
- Department of Clinical Laboratory Diagnostics, Medical University of Bialystok, Waszyngtona 15A, 15-269 Bialystok, Poland
| |
Collapse
|
79
|
Creatine promotes cancer metastasis through activation of Smad2/3. Cell Metab 2021; 33:1111-1123.e4. [PMID: 33811821 DOI: 10.1016/j.cmet.2021.03.009] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 02/15/2021] [Accepted: 03/11/2021] [Indexed: 12/24/2022]
Abstract
As one of the most popular nutrient supplements, creatine has been highly used to increase muscle mass and improve exercise performance. Here, we report an adverse effect of creatine using orthotopic mouse models, showing that creatine promotes colorectal and breast cancer metastasis and shortens mouse survival. We show that glycine amidinotransferase (GATM), the rate-limiting enzyme for creatine synthesis, is upregulated in liver metastases. Dietary uptake, or GATM-mediated de novo synthesis of creatine, enhances cancer metastasis and shortens mouse survival by upregulation of Snail and Slug expression via monopolar spindle 1 (MPS1)-activated Smad2 and Smad3 phosphorylation. GATM knockdown or MPS1 inhibition suppresses cancer metastasis and benefits mouse survival by downregulating Snail and Slug. Our findings call for using caution when considering dietary creatine to improve muscle mass or treat diseases and suggest that targeting GATM or MPS1 prevents cancer metastasis, especially metastasis of transforming growth factor beta receptor mutant colorectal cancers.
Collapse
|
80
|
The effect of platelet G proteins on platelet extravasation and tumor growth in the murine model of ovarian cancer. Blood Adv 2021; 5:1947-1951. [PMID: 33821990 DOI: 10.1182/bloodadvances.2020003410] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/02/2021] [Indexed: 12/21/2022] Open
Abstract
We and other investigators have shown that platelets promote metastasis and the growth of tumors. Our rationale for conducting this study is that platelets' prometastatic and progrowth effects depend on a close encounter between platelets and cancer cells. This interaction occurs inside blood vessels with circulating tumor cells and outside blood vessels with cancer cells residing in the tumor parenchyma. Our hypothesis was that platelet extravasation is required for the effect of platelets on tumor growth. Platelets respond to environmental stimuli by activation of G protein-coupled receptors on their surface. We investigated the impact of various platelet G proteins on the growth of ovarian cancer tumors and platelet extravasation. We used mice with platelet-specific deficiency of Gαi2 (Gi), Gα13 (G13), or Gαq (Gq) in a syngeneic ovarian cancer model. We measured the total weight of tumor nodules resected from tumor-bearing mice. We developed methods for automated whole-slide image acquisition and unbiased computerized image analysis to quantify extravasated platelets. We compared the number of platelets inside tumor nodules of platelet G protein-deficient tumor-bearing mice. We found that deficiency of Gi and G13, but not Gq, in platelets resulted in smaller tumors compared with those in corresponding littermates. Deficiency of Gi and G13 in platelets reduced the number of extravasated platelets by >90%, but deficiency of Gq did not reduce the number of extravasated platelets significantly. The lack of Gi or G13 in platelets reduced platelet extravasation into the tumor and tumor growth.
Collapse
|
81
|
Abstract
Until recently, the nucleic acid content of platelets was considered to be fully determined by their progenitor megakaryocyte. However, it is now well understood that additional mediators (eg, cancer cells) can intervene, thereby influencing the RNA repertoire of platelets. Platelets are highly dynamic cells that are able to communicate and influence their environment. For instance, platelets have been involved in various steps of cancer development and progression by supporting tumor growth, survival, and dissemination. Cancer cells can directly and/or indirectly influence platelet RNA content, resulting in tumor-mediated "education" of platelets. Alterations in the tumor-educated platelet RNA profile have been described as a novel source of potential biomarkers. Individual platelet RNA biomarkers as well as complex RNA signatures may be used for early detection of cancer and treatment monitoring. Here, we review the RNA transfer occurring between cancer cells and platelets. We explore the potential use of platelet RNA biomarkers as a liquid biopsy biosource and discuss methods to evaluate the transcriptomic content of platelets.
Collapse
|
82
|
Cacic D, Reikvam H, Nordgård O, Meyer P, Hervig T. Platelet Microparticles Protect Acute Myelogenous Leukemia Cells against Daunorubicin-Induced Apoptosis. Cancers (Basel) 2021; 13:cancers13081870. [PMID: 33919720 PMCID: PMC8070730 DOI: 10.3390/cancers13081870] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/08/2021] [Accepted: 04/11/2021] [Indexed: 12/21/2022] Open
Abstract
The role of platelets in cancer development and progression is increasingly evident, and several platelet-cancer interactions have been discovered, including the uptake of platelet microparticles (PMPs) by cancer cells. PMPs inherit a myriad of proteins and small RNAs from the parental platelets, which in turn can be transferred to cancer cells following internalization. However, the exact effect this may have in acute myelogenous leukemia (AML) is unknown. In this study, we sought to investigate whether PMPs could transfer their contents to the THP-1 cell line and if this could change the biological behavior of the recipient cells. Using acridine orange stained PMPs, we demonstrated that PMPs were internalized by THP-1 cells, which resulted in increased levels of miR-125a, miR-125b, and miR-199. In addition, co-incubation with PMPs protected THP-1 and primary AML cells against daunorubicin-induced cell death. We also showed that PMPs impaired cell growth, partially inhibited cell cycle progression, decreased mitochondrial membrane potential, and induced differentiation toward macrophages in THP-1 cells. Our results suggest that this altering of cell phenotype, in combination with decrease in cell activity may offer resistance to daunorubicin-induced apoptosis, as serum starvation also yielded a lower frequency of dead and apoptotic cells when treated with daunorubicin.
Collapse
Affiliation(s)
- Daniel Cacic
- Department of Hematology and Oncology, Stavanger University Hospital, 4068 Stavanger, Norway; (O.N.); (P.M.)
- Correspondence:
| | - Håkon Reikvam
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (H.R.); (T.H.)
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Oddmund Nordgård
- Department of Hematology and Oncology, Stavanger University Hospital, 4068 Stavanger, Norway; (O.N.); (P.M.)
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4036 Stavanger, Norway
| | - Peter Meyer
- Department of Hematology and Oncology, Stavanger University Hospital, 4068 Stavanger, Norway; (O.N.); (P.M.)
| | - Tor Hervig
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (H.R.); (T.H.)
- Laboratory of Immunology and Transfusion Medicine, Haugesund Hospital, 5528 Haugesund, Norway
| |
Collapse
|
83
|
Ward MP, E Kane L, A Norris L, Mohamed BM, Kelly T, Bates M, Clarke A, Brady N, Martin CM, Brooks RD, Brooks DA, Selemidis S, Hanniffy S, Dixon EP, A O'Toole S, J O'Leary J. Platelets, immune cells and the coagulation cascade; friend or foe of the circulating tumour cell? Mol Cancer 2021; 20:59. [PMID: 33789677 PMCID: PMC8011144 DOI: 10.1186/s12943-021-01347-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer cells that transit from primary tumours into the circulatory system are known as circulating tumour cells (CTCs). These cancer cells have unique phenotypic and genotypic characteristics which allow them to survive within the circulation, subsequently extravasate and metastasise. CTCs have emerged as a useful diagnostic tool using "liquid biopsies" to report on the metastatic potential of cancers. However, CTCs by their nature interact with components of the blood circulatory system on a constant basis, influencing both their physical and morphological characteristics as well as metastatic capabilities. These properties and the associated molecular profile may provide critical diagnostic and prognostic capabilities in the clinic. Platelets interact with CTCs within minutes of their dissemination and are crucial in the formation of the initial metastatic niche. Platelets and coagulation proteins also alter the fate of a CTC by influencing EMT, promoting pro-survival signalling and aiding in evading immune cell destruction. CTCs have the capacity to directly hijack immune cells and utilise them to aid in CTC metastatic seeding processes. The disruption of CTC clusters may also offer a strategy for the treatment of advance staged cancers. Therapeutic disruption of these heterotypical interactions as well as direct CTC targeting hold great promise, especially with the advent of new immunotherapies and personalised medicines. Understanding the molecular role that platelets, immune cells and the coagulation cascade play in CTC biology will allow us to identify and characterise the most clinically relevant CTCs from patients. This will subsequently advance the clinical utility of CTCs in cancer diagnosis/prognosis.
Collapse
Affiliation(s)
- Mark P Ward
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland.
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland.
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland.
| | - Laura E Kane
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Lucy A Norris
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
- Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin 8, Ireland
| | - Bashir M Mohamed
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Tanya Kelly
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Mark Bates
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Andres Clarke
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Nathan Brady
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Cara M Martin
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| | - Robert D Brooks
- Cancer Research Institute, University of South Australia, 5001, Adelaide, Australia
| | - Doug A Brooks
- Cancer Research Institute, University of South Australia, 5001, Adelaide, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, RMIT University, Victoria, 3083, Bundoora, Australia
| | | | - Eric P Dixon
- BD Technologies and Innovation, Research Triangle Park, NC, USA
| | - Sharon A O'Toole
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
- Department of Obstetrics and Gynaecology, Trinity College Dublin, Dublin 8, Ireland
| | - John J O'Leary
- Department of Histopathology and Morbid Anatomy, Trinity College Dublin, Dublin 8, Ireland
- Emer Casey Molecular Pathology Research Laboratory, Coombe Women and Infants University Hospital, Dublin 8, Ireland
- Trinity St. James's Cancer Institute, St James's Hospital, Dublin 8, Ireland
| |
Collapse
|
84
|
Sier VQ, de Vries MR, van der Vorst JR, Vahrmeijer AL, van Kooten C, Cruz LJ, de Geus-Oei LF, Ferreira V, Sier CFM, Alves F, Muthana M. Cell-Based Tracers as Trojan Horses for Image-Guided Surgery. Int J Mol Sci 2021; 22:E755. [PMID: 33451116 PMCID: PMC7828607 DOI: 10.3390/ijms22020755] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 12/29/2020] [Accepted: 12/30/2020] [Indexed: 12/14/2022] Open
Abstract
Surgeons rely almost completely on their own vision and palpation to recognize affected tissues during surgery. Consequently, they are often unable to distinguish between different cells and tissue types. This makes accurate and complete resection cumbersome. Targeted image-guided surgery (IGS) provides a solution by enabling real-time tissue recognition. Most current targeting agents (tracers) consist of antibodies or peptides equipped with a radiolabel for Positron Emission Tomography (PET) and Single Photon Emission Computed Tomography (SPECT), magnetic resonance imaging (MRI) labels, or a near-infrared fluorescent (NIRF) dye. These tracers are preoperatively administered to patients, home in on targeted cells or tissues, and are visualized in the operating room via dedicated imaging systems. Instead of using these 'passive' tracers, there are other, more 'active' approaches of probe delivery conceivable by using living cells (macrophages/monocytes, neutrophils, T cells, mesenchymal stromal cells), cell(-derived) fragments (platelets, extracellular vesicles (exosomes)), and microorganisms (bacteria, viruses) or, alternatively, 'humanized' nanoparticles. Compared with current tracers, these active contrast agents might be more efficient for the specific targeting of tumors or other pathological tissues (e.g., atherosclerotic plaques). This review provides an overview of the arsenal of possibilities applicable for the concept of cell-based tracers for IGS.
Collapse
Affiliation(s)
- Vincent Q. Sier
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (M.R.d.V.); (J.R.v.d.V.); (A.L.V.)
| | - Margreet R. de Vries
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (M.R.d.V.); (J.R.v.d.V.); (A.L.V.)
| | - Joost R. van der Vorst
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (M.R.d.V.); (J.R.v.d.V.); (A.L.V.)
| | - Alexander L. Vahrmeijer
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (M.R.d.V.); (J.R.v.d.V.); (A.L.V.)
| | - Cornelis van Kooten
- Department of Nephrology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Luis J. Cruz
- Department of Radiology, Translational Nanomaterials and Imaging Group, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, 2300 RC Leiden, The Netherlands;
- Biomedical Photonic Imaging Group, University of Twente, 7522 NB Enschede, The Netherlands
| | - Valerie Ferreira
- Department of Research and Development, UniQure, 1100 DA Amsterdam, The Netherlands;
| | - Cornelis F. M. Sier
- Department of Surgery, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (V.Q.S.); (M.R.d.V.); (J.R.v.d.V.); (A.L.V.)
- Percuros B.V. Leiden, 2333 CL Leiden, The Netherlands
| | - Frauke Alves
- Translational Molecular Imaging, Clinic of Hematology and Medical Oncology, Institute of Diagnostic and Interventional Radiology, University Medicine Center Göttingen and Max-Planck-Institute for Experimental Medicine, 37075 Göttingen, Germany;
| | - Munitta Muthana
- Department of Infection and Immunity, University of Sheffield, Sheffield S10 2RX, UK;
| |
Collapse
|
85
|
Bouck EG, de la Fuente M, Zunica ER, Li W, Mumaw MM, Nieman MT. Murine cadherin-6 mediates thrombosis in vivo in a platelet-independent manner. Res Pract Thromb Haemost 2021; 5:125-131. [PMID: 33537536 PMCID: PMC7845066 DOI: 10.1002/rth2.12458] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 10/26/2020] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Platelet adhesion is the critical process mediating stable thrombus formation. Previous reports of cadherin-6 on human platelets have demonstrated its role in platelet aggregation and thrombus formation. OBJECTIVES We aimed to further characterize the importance of cadherin-6 in thrombosis in vivo. METHODS Cadherin-6 platelet expression was evaluated by western blotting, flow cytometry, and immunoprecipitation. Thrombosis was evaluated using the FeCl3 and Rose Bengal carotid artery models in C57Bl6 mice treated with anti-cadherin-6 or IgG and wild-type or Cdh6-/- mice. Platelet function was compared in wild-type and Cdh6-/- mice using tail-clip assays, aggregometry, and flow cytometry. RESULTS Human platelet expression of cadherin-6 was confirmed at ~3000 copies per platelet. Cdh6-/- mice or those treated with anti-cadherin-6 antibody showed an increased time to occlusion in both thrombosis models. Cadherin-6 was not expressed on mouse platelets, and there were no differences in tail bleeding times, platelet aggregation, or platelet activation in wild-type versus Cdh6-/- mice. CONCLUSIONS Cadherin-6 plays an essential role in thrombosis in vivo. However, cadherin-6 is not expressed on murine platelets. These data are in contrast to human platelets, which express a functional cadherin-6/catenin complex. The essential, platelet-independent role for cadherin-6 in hemostasis may allow it to be an effective and safe therapeutic target.
Collapse
Affiliation(s)
- Emma G. Bouck
- Department of PharmacologyCase Western Reserve UniversityClevelandOHUSA
| | | | | | - Wei Li
- Deparmtent of Biomedical SciencesMarshall University Joan C. Edwards School of MedicineHuntingtonWVUSA
| | - Michele M. Mumaw
- Department of PharmacologyCase Western Reserve UniversityClevelandOHUSA
| | - Marvin T. Nieman
- Department of PharmacologyCase Western Reserve UniversityClevelandOHUSA
| |
Collapse
|
86
|
Pu F, Li X, Wang S, Huang Y, Wang D. Platelet supernatant with longer storage inhibits tumor cell growth. Transfus Apher Sci 2020; 60:103042. [PMID: 33376060 DOI: 10.1016/j.transci.2020.103042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Platelet transfusion is an essential supportive treatment for cancer patients. Platelets promote metastasis, but their role in tumor growth remains controversial. This study aimed to explore the impact of apheresis platelet supernatants of different storage periods on tumor cell growth, optimize blood transfusion timing, and provide a reference for reducing the risks of platelet transfusion in cancer patients. METHODS Eight human tumor cell lines, including HepG2, HuH7, SMMC-7721, HeLa, HCT116, MCF-7, K562, and Jurkat were cultured. After culturing the platelet supernatant of days 0, 3, 5, and 7 with tumor cells, counting kit 8 (CCK-8), scratch assay, and propidium iodide (PI) were used to evaluate cell proliferation, migration, and cell cycle, respectively. Metabolomics analysis was performed to confirm whether differential metabolites produced during platelet storage are involved in the cancer pathway. RESULTS Platelet supernatants inhibit tumor cell proliferation by blocking the cell cycle at the G0/G1 phase, and their inhibitory effect increases with storage time. However, platelets promote tumor cells to form cytoskeletal connections, thereby promoting migration. Besides, metabonomics analysis of platelet supernatants during different storage periods reveals that upregulated differential metabolites are involved in cancer-related pathways. CONCLUSION The role of platelets in tumor cells is two-sided, that is, they inhibit proliferation while promoting migration. Therefore, additional in-depth studies on the appropriate timing of platelet transfusion are necessary.
Collapse
Affiliation(s)
- Fei Pu
- Department of Blood Transfusion, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China; Department of Blood Transfusion, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xiaofei Li
- Department of Blood Transfusion, Beijing Friendship Hospital, Capital Medical University, Beijing, China; Department of Blood Transfusion, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Shufang Wang
- Department of Blood Transfusion, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yuanshuai Huang
- Department of Blood Transfusion, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China.
| | - Deqing Wang
- Department of Blood Transfusion, The First Medical Center, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
87
|
Impact of Epicatechin on the Procoagulant Activities of Microparticles. Nutrients 2020; 12:nu12102935. [PMID: 32992756 PMCID: PMC7601556 DOI: 10.3390/nu12102935] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/23/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023] Open
Abstract
Microparticles play a role in cardiovascular disease pathology. The flavanol-like epicatechin is increasingly considered due to its cardioprotective effects. The aim of this study was to investigate the impact of epicatechin on microparticle generation, phenotype and procoagulant properties. Plasma samples from 15 healthy subjects were incubated with increasing concentrations of epicatechin (1 to 100 μM). Then, the expression of glycoprotein IIb, phosphatidylserine (PS), glycoprotein Ib (GPIb) and P-selectin was assessed by flow cytometry analysis after (or not) platelet stimulation. Microparticle procoagulant activity was determined using ZymuphenTM MP and ZymuphenTM MP-TF for phospholipid and tissue factor content, and with thrombin generation (TG) assays for procoagulant function. Platelet microparticles that express GPIb (/µL) decreased from 20,743 ± 24,985 (vehicle) to 14,939 ± 14,333 (p = 0.6), 21,366 ± 16,949 (p = 0.9) and 15,425 ± 9953 (p < 0.05) in samples incubated with 1, 10 and 100 µM epicatechin, respectively. Microparticle concentration (nM PS) decreased from 5.6 ± 2.0 (vehicle) to 5.1 ± 2.2 (p = 0.5), 4.5 ± 1.5 (p < 0.05) and 4.7 ± 2.0 (p < 0.05) in samples incubated with 1, 10 and 100µM epicatechin, respectively. Epicatechin had no impact on tissue factor-positive microparticle concentration. Epicatechin decreased TG (endogenous thrombin potential, nM.min) from 586 ± 302 to 509 ± 226 (p = 0.3), 512 ± 270 (p = 0.3) and 445 ± 283 (p < 0.05). These findings indicate that epicatechin affects microparticle release, phenotype and procoagulant properties.
Collapse
|
88
|
Hyslop SR, Alexander M, Thai AA, Kersbergen A, Kueh AJ, Herold MJ, Corbin J, Gangatirkar P, Ng AP, Solomon BJ, Alexander WS, Sutherland KD, Josefsson EC. Targeting platelets for improved outcome in KRAS-driven lung adenocarcinoma. Oncogene 2020; 39:5177-5186. [PMID: 32535617 DOI: 10.1038/s41388-020-1357-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 06/02/2020] [Accepted: 06/04/2020] [Indexed: 12/21/2022]
Abstract
Elevated platelet count is associated with poor survival in certain solid cancers, including lung cancer. In addition, experimental transplantation of cancer cell lines has uncovered a role for platelets in blood-borne metastasis. These studies, however, do not account for heterogeneity between lung cancer subtypes. Subsequently, the role of platelets in the major subtypes of non-small cell lung cancer (adenocarcinoma (ADC) and squamous cell carcinoma (SqCC)) is not fully understood. We utilised an autochthonous KrasLSL-G12D/+;p53flox/flox mouse model of lung ADC together with genetic models of thrombocytopenia to interrogate the role of platelets in lung cancer growth and progression. While thrombocytopenia failed to impact primary tumour growth, in experimental metastatic models however, thrombocytopenic mice displayed significantly extended survival. Utilising a novel thrombocytopenic immunocompromised mouse, the importance of platelets in metastatic dissemination was confirmed with human KRAS-mutant ADC cell lines. Finally, retrospective analysis of a NSCLC patient cohort revealed thrombocytosis was predictive of poor survival in ADC patients with metastatic disease. Interestingly, this association was not apparent in SqCC patients. Overall, these data highlight the possibility of patient stratification using thrombocytosis as a biomarker, and indicates opportunities for potential novel treatment strategies that combine anti-platelet and lung cancer therapies.
Collapse
Affiliation(s)
- Stephanie R Hyslop
- ACRF Cancer Biology & Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Marliese Alexander
- Pharmacy Department, Peter MacCallum Cancer Centre, University of Melbourne, Parkville, VIC, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia
| | - Alesha A Thai
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Ariena Kersbergen
- ACRF Cancer Biology & Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Andrew J Kueh
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia.,Blood Cells & Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Marco J Herold
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia.,Blood Cells & Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Jason Corbin
- ACRF Cancer Biology & Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Pradnya Gangatirkar
- ACRF Cancer Biology & Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Ashley P Ng
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia.,Blood Cells & Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Benjamin J Solomon
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC, Australia.,Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Warren S Alexander
- Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia.,Blood Cells & Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Kate D Sutherland
- ACRF Cancer Biology & Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Emma C Josefsson
- ACRF Cancer Biology & Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia. .,Department of Medical Biology, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
89
|
Wei CY, Zhu MX, Lu NH, Liu JQ, Yang YW, Zhang Y, Shi YD, Feng ZH, Li JX, Qi FZ, Gu JY. Circular RNA circ_0020710 drives tumor progression and immune evasion by regulating the miR-370-3p/CXCL12 axis in melanoma. Mol Cancer 2020; 19:84. [PMID: 32381016 PMCID: PMC7204052 DOI: 10.1186/s12943-020-01191-9] [Citation(s) in RCA: 115] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/24/2020] [Indexed: 02/26/2023] Open
Abstract
Background Circular RNAs (circRNAs) have been reported to have critical regulatory roles in tumor biology. However, their contribution to melanoma remains largely unknown. Methods CircRNAs derived from oncogene CD151 were detected and verified by analyzing a large number of melanoma samples through quantitative real-time polymerase chain reaction (qRT-PCR). Melanoma cells were stably transfected with lentiviruses using circ_0020710 interference or overexpression plasmid, and then CCK-8, colony formation, wound healing, transwell invasion assays, and mouse xenograft models were employed to assess the potential role of circ_0020710. RNA immunoprecipitation, luciferase reporter assay and fluorescence in situ hybridization were used to evaluate the underlying mechanism of circ_0020710. Results Our findings indicated that circ_0020710 was generally overexpressed in melanoma tissues, and high level of circ_0020710 was positively correlated with malignant phenotype and poor prognosis of melanoma patients. Elevated circ_0020710 promoted melanoma cell proliferation, migration and invasion in vitro as well as tumor growth in vivo. Mechanistically, we found that high level of circ_0020710 could upregulate the CXCL12 expression via sponging miR-370-3p. CXCL12 downregulation could reverse the malignant behavior of melanoma cells conferred by circ_0020710 over expression. Moreover, we also found that elevated circ_0020710 was correlated with cytotoxic lymphocyte exhaustion, and a combination of AMD3100 (the CXCL12/CXCR4 axis inhibitor) and anti-PD-1 significantly attenuated tumor growth. Conclusions Elevated circ_0020710 drives tumor progression via the miR-370-3p/CXCL12 axis, and circ_0020710 is a potential target for melanoma treatment.
Collapse
Affiliation(s)
- Chuan-Yuan Wei
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Meng-Xuan Zhu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Nan-Hang Lu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Jia-Qi Liu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Yan-Wen Yang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Yong Zhang
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Yue-Dong Shi
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Zi-Hao Feng
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Jia-Xia Li
- Department of Neurology, Hefei Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230000, P. R. China
| | - Fa-Zhi Qi
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.
| | - Jian-Ying Gu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China.
| |
Collapse
|
90
|
The endothelial barrier and cancer metastasis: Does the protective facet of platelet function matter? Biochem Pharmacol 2020; 176:113886. [PMID: 32113813 DOI: 10.1016/j.bcp.2020.113886] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/24/2020] [Indexed: 12/16/2022]
Abstract
Overwhelming evidence suggests that platelets have a detrimental role in promoting cancer spread via platelet-cancer cell interactions linked to thrombotic mechanisms. On the other hand, a beneficial role of platelets in the preservation of the endothelial barrier in inflammatory conditions has been recently described, a phenomenon that could also operate in cancer-related inflammation. It is tempting to speculate that some antiplatelet strategies to combat cancer metastasis may impair the endogenous platelet-dependent mechanisms preserving endothelial barrier function. If the protective function of platelets is impaired, it may lead to increased endothelial permeability and more efficient cancer cell intravasation in the primary tumor and cancer cell extravasation at metastatic sites. In this commentary, we discuss current evidence that could support this hypothesis.
Collapse
|
91
|
Jia G, Kong J, Yao C, Wu S, Sun W. Platelet lysates in Hepatocellular Carcinoma patients after radiofrequency ablation facilitate tumor proliferation, invasion and vasculogenic mimicry. Int J Med Sci 2020; 17:2104-2112. [PMID: 32922171 PMCID: PMC7484666 DOI: 10.7150/ijms.44405] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 07/06/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Platelets play important roles in tumorigenesis, angiogenesis and metastatic dissemination of tumor cells. Radiofrequency ablation (RFA) could increase the circulating tumor cells in patients with primary or metastatic lung tumors. Whether platelet lysates in hepatocellular carcinoma (HCC) after RFA promote tumor progression has not been elaborated. Methods: HCC patients within Milan Criteria and without taking anti-platelet drugs were selected in the study. MTT assay, colony formation assay, transwell assay, tube formation and western blot were used to evaluate the effect of platelet lysates on HCC cells in vitro. Lung metastatic assay was performed in vivo. Results: Platelet lysates from patients after RFA promoted cell proliferation, colony formation, migration, invasion and vasculogenic mimicry in Hep3B and HCCLM3 cells compared with those from patients before RFA. Platelet lysates after RFA significantly increased the expression of p-Akt, p-Smad3 and snail, and decreased the expression of E-cadherin compared with those before RFA in Hep3B and HCCLM3 cells. Hep3B-Luc2-tdT cells incubation with platelet lysates from patients after RFA displayed enhanced lung metastasis compared with those before RFA. Conclusions: Platelet lysates from HCC patients after RFA promoted the proliferation, migration, invasion and vasculogenic mimicry of HCC cells, which indicated that RFA in combination with anti-platelet drug may be used to improve the prognosis of HCC.
Collapse
Affiliation(s)
- Guoqun Jia
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
| | - Jian Kong
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
| | - Changyu Yao
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
| | - Shilun Wu
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
| | - Wenbing Sun
- Department of Hepatobiliary Surgery, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|