1
|
Sureka N, Zaheer S. Regulatory T Cells in Tumor Microenvironment: Therapeutic Approaches and Clinical Implications. Cell Biol Int 2025. [PMID: 40365758 DOI: 10.1002/cbin.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/19/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025]
Abstract
Regulatory T cells (Tregs), previously referred to as suppressor T cells, represent a distinct subset of CD4+ T cells that are uniquely specialized for immune suppression. They are characterized by the constitutive expression of the transcription factor FoxP3 in their nuclei, along with CD25 (the IL-2 receptor α-chain) and CTLA-4 on their cell surface. Tregs not only restrict natural killer cell-mediated cytotoxicity but also inhibit the proliferation of CD4+ and CD8+ T-cells and suppress interferon-γ secretion by immune cells, ultimately impairing an effective antitumor immune response. Treg cells are widely recognized as a significant barrier to the effectiveness of tumor immunotherapy in clinical settings. Extensive research has consistently shown that Treg cells play a pivotal role in facilitating tumor initiation and progression. Conversely, the depletion of Treg cells has been linked to a marked delay in tumor growth and development.
Collapse
Affiliation(s)
- Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
2
|
Xu H, Ma H, Li Y, Bi S, Cai K, Wu L, Zhang L, Guan H, Li C, Yang J, Qiu P. Propylene glycol alginate sodium sulfate suppressed lung metastasis by blocking P-selectin to recruit CD4 regulatory T cells. Int J Biol Macromol 2024; 279:134976. [PMID: 39179086 DOI: 10.1016/j.ijbiomac.2024.134976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/12/2024] [Accepted: 08/21/2024] [Indexed: 08/26/2024]
Abstract
P-selectin has been shown to enhance growth and metastasis of mouse tumors by promoting regulatory T cell (Treg) infiltration into the tumors. Theoretically, a P-selectin antagonist could suppress the process. Popylene glycol alginate sodium sulfate (PSS) is a heparin-like marine drug, which was originally approved to treat cardiovascular disease in China. Previously, we reported that PSS was an effective P-selectin antagonist in vitro. However, it is unknown whether PSS can regulate Treg infiltration and its effect on lung metastasis in vivo. Our results showed that PSS at 30 mg/kg significantly suppressed lung metastasis and improved overall survival, with potency comparable to the positive control LMWH. Mechanistic study indicated that PSS blocked tumor cells adhesion and activated platelets by directly binding with activated platelet's P-selectin. Compared to the model group, PSS decreased the percent of Tregs by 63 % in lungs after treating for 21 days while increasing CD8+ T cells (1.59-fold) and Granzyme B+ CD8 T cells (2.08-fold)' percentage for generating an adaptive response for systemic tumor suppression. The study indicated that the P-selectin antagonist, PSS, suppressed lung metastasis by inhibiting the infiltration of regulatory T cells (Treg) into the tumors.
Collapse
Affiliation(s)
- Huixin Xu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China; Marine Biomedical Research Institute of Qiangdao, 23 Hongkong East Rd, Qingdao, Shandong 266003, China
| | - He Ma
- College of Veterinary Medicine, Qingdao Agricultural University, 700 Changcheng Rd, Qingdao, Shandong,266109, China
| | - Yannan Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China
| | - Shijie Bi
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China
| | - Kaiyu Cai
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China
| | - Lijuan Wu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China
| | - Lei Zhang
- Research Center of Traditional Chinese Medicine and Clinical Pharmacy, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250014, China
| | - Huashi Guan
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China; Marine Biomedical Research Institute of Qiangdao, 23 Hongkong East Rd, Qingdao, Shandong 266003, China
| | - Chunxia Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China
| | - Jinbo Yang
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China; Marine Biomedical Research Institute of Qiangdao, 23 Hongkong East Rd, Qingdao, Shandong 266003, China
| | - Peiju Qiu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Rd, Qingdao, Shandong 266003, China; Marine Biomedical Research Institute of Qiangdao, 23 Hongkong East Rd, Qingdao, Shandong 266003, China
| |
Collapse
|
3
|
Shi X, Wang X, Yao W, Shi D, Shao X, Lu Z, Chai Y, Song J, Tang W, Wang X. Mechanism insights and therapeutic intervention of tumor metastasis: latest developments and perspectives. Signal Transduct Target Ther 2024; 9:192. [PMID: 39090094 PMCID: PMC11294630 DOI: 10.1038/s41392-024-01885-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 05/29/2024] [Accepted: 06/10/2024] [Indexed: 08/04/2024] Open
Abstract
Metastasis remains a pivotal characteristic of cancer and is the primary contributor to cancer-associated mortality. Despite its significance, the mechanisms governing metastasis are not fully elucidated. Contemporary findings in the domain of cancer biology have shed light on the molecular aspects of this intricate process. Tumor cells undergoing invasion engage with other cellular entities and proteins en route to their destination. Insights into these engagements have enhanced our comprehension of the principles directing the movement and adaptability of metastatic cells. The tumor microenvironment plays a pivotal role in facilitating the invasion and proliferation of cancer cells by enabling tumor cells to navigate through stromal barriers. Such attributes are influenced by genetic and epigenetic changes occurring in the tumor cells and their surrounding milieu. A profound understanding of the metastatic process's biological mechanisms is indispensable for devising efficacious therapeutic strategies. This review delves into recent developments concerning metastasis-associated genes, important signaling pathways, tumor microenvironment, metabolic processes, peripheral immunity, and mechanical forces and cancer metastasis. In addition, we combine recent advances with a particular emphasis on the prospect of developing effective interventions including the most popular cancer immunotherapies and nanotechnology to combat metastasis. We have also identified the limitations of current research on tumor metastasis, encompassing drug resistance, restricted animal models, inadequate biomarkers and early detection methods, as well as heterogeneity among others. It is anticipated that this comprehensive review will significantly contribute to the advancement of cancer metastasis research.
Collapse
Affiliation(s)
- Xiaoli Shi
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
- School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Xinyi Wang
- The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wentao Yao
- Department of Urology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, Jiangsu, China
| | - Dongmin Shi
- Department of Medical Oncology, Shanghai Changzheng Hospital, Shanghai, China
| | - Xihuan Shao
- The Fourth Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhengqing Lu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Yue Chai
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Jinhua Song
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China.
| | - Weiwei Tang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China.
| | - Xuehao Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences; NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China.
- School of Medicine, Southeast University, Nanjing, Jiangsu, China.
| |
Collapse
|
4
|
Rabi LT, Valente DZ, de Souza Teixeira E, Peres KC, de Oliveira Almeida M, Bufalo NE, Ward LS. Potential new cancer biomarkers revealed by quantum chemistry associated with bioinformatics in the study of selectin polymorphisms. Heliyon 2024; 10:e28830. [PMID: 38586333 PMCID: PMC10998122 DOI: 10.1016/j.heliyon.2024.e28830] [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: 10/17/2023] [Revised: 03/22/2024] [Accepted: 03/26/2024] [Indexed: 04/09/2024] Open
Abstract
Understanding the complex mechanisms involved in diseases caused by or related to important genetic variants has led to the development of clinically useful biomarkers. However, the increasing number of described variants makes it difficult to identify variants worthy of investigation, and poses challenges to their validation. We combined publicly available datasets and open source robust bioinformatics tools with molecular quantum chemistry methods to investigate the involvement of selectins, important molecules in the cell adhesion process that play a fundamental role in the cancer metastasis process. We applied this strategy to investigate single nucleotide variants (SNPs) in the intronic and UTR regions and missense SNPs with amino acid changes in the SELL, SELP, SELE, and SELPLG genes. We then focused on thyroid cancer, seeking these SNPs potential to identify biomarkers for susceptibility, diagnosis, prognosis, and therapeutic targets. We demonstrated that SELL gene polymorphisms rs2229569, rs1131498, rs4987360, rs4987301 and rs2205849; SELE gene polymorphisms rs1534904 and rs5368; rs3917777, rs2205894 and rs2205893 of SELP gene; and rs7138370, rs7300972 and rs2228315 variants of SELPLG gene may produce important alterations in the DNA structure and consequent changes in the morphology and function of the corresponding proteins. In conclusion, we developed a strategy that may save valuable time and resources in future investigations, as we were able to provide a solid foundation for the selection of selectin gene variants that may become important biomarkers and deserve further investigation in cancer patients. Large-scale clinical studies in different ethnic populations and laboratory experiments are needed to validate our results.
Collapse
Affiliation(s)
- Larissa Teodoro Rabi
- Laboratory of Cancer Molecular Genetics, Faculty of Medical Sciences, State University of Campinas (UNI-CAMP), Campinas, SP, Brazil
- .Department of Biomedicine, Nossa Senhora do Patrocínio University Center (CEUNSP), Itu, SP, Brazil
- Institute of Health Sciences, Paulista University (UNIP), Campinas, SP, Brazil
| | - Davi Zanoni Valente
- Laboratory of Cancer Molecular Genetics, Faculty of Medical Sciences, State University of Campinas (UNI-CAMP), Campinas, SP, Brazil
| | - Elisangela de Souza Teixeira
- Laboratory of Cancer Molecular Genetics, Faculty of Medical Sciences, State University of Campinas (UNI-CAMP), Campinas, SP, Brazil
| | - Karina Colombera Peres
- Laboratory of Cancer Molecular Genetics, Faculty of Medical Sciences, State University of Campinas (UNI-CAMP), Campinas, SP, Brazil
- Department of Medicine, Max Planck University Center, Campinas, SP, Brazil
| | | | - Natassia Elena Bufalo
- Laboratory of Cancer Molecular Genetics, Faculty of Medical Sciences, State University of Campinas (UNI-CAMP), Campinas, SP, Brazil
- Department of Medicine, Max Planck University Center, Campinas, SP, Brazil
- Department of Medicine, São Leopoldo Mandic and Research Center, Campinas, SP, Brazil
| | - Laura Sterian Ward
- Laboratory of Cancer Molecular Genetics, Faculty of Medical Sciences, State University of Campinas (UNI-CAMP), Campinas, SP, Brazil
| |
Collapse
|
5
|
Muluh TA, Shu XS, Ying Y. Targeting cancer metabolic vulnerabilities for advanced therapeutic efficacy. Biomed Pharmacother 2023; 162:114658. [PMID: 37031495 DOI: 10.1016/j.biopha.2023.114658] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/11/2023] Open
Abstract
Cancer metabolism is how cancer cells utilize nutrients and energy to support their growth and proliferation. Unlike normal cells, cancer cells have a unique metabolic profile that allows them to generate energy and the building blocks they need for rapid growth and division. This metabolic profile is marked by an increased reliance on glucose and glutamine as energy sources and changes in how cancer cells use and make key metabolic intermediates like ATP, NADH, and NADPH. This script analyzes a comprehensive overview of the latest advances in tumor metabolism, identifying the key unresolved issues, elaborates on how tumor cells differ from normal cells in their metabolism of nutrients, and explains how tumor cells conflate growth signals and nutrients to proliferate. The metabolic interaction of tumorigenesis and lipid metabolism within the tumor microenvironment and the role of ROS as an anti-tumor agent by mediating various signaling pathways for clinical cancer therapeutic targeting are outlined. Cancer metabolism is highly dynamic and heterogeneous; thus, advanced technologies to better investigate metabolism at the unicellular level without altering tumor tissue are necessary for better research and clinical transformation. The study of cancer metabolism is an area of active research, as scientists seek to understand the underlying metabolic changes that drive cancer growth and to identify potential therapeutic targets.
Collapse
Affiliation(s)
- Tobias Achu Muluh
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Xing-Sheng Shu
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Ying Ying
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China; Marshall Laboratory of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
6
|
Interactions between Platelets and Tumor Microenvironment Components in Ovarian Cancer and Their Implications for Treatment and Clinical Outcomes. Cancers (Basel) 2023; 15:cancers15041282. [PMID: 36831623 PMCID: PMC9953912 DOI: 10.3390/cancers15041282] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/07/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023] Open
Abstract
Platelets, the primary operatives of hemostasis that contribute to blood coagulation and wound healing after blood vessel injury, are also involved in pathological conditions, including cancer. Malignancy-associated thrombosis is common in ovarian cancer patients and is associated with poor clinical outcomes. Platelets extravasate into the tumor microenvironment in ovarian cancer and interact with cancer cells and non-cancerous elements. Ovarian cancer cells also activate platelets. The communication between activated platelets, cancer cells, and the tumor microenvironment is via various platelet membrane proteins or mediators released through degranulation or the secretion of microvesicles from platelets. These interactions trigger signaling cascades in tumors that promote ovarian cancer progression, metastasis, and neoangiogenesis. This review discusses how interactions between platelets, cancer cells, cancer stem cells, stromal cells, and the extracellular matrix in the tumor microenvironment influence ovarian cancer progression. It also presents novel potential therapeutic approaches toward this gynecological cancer.
Collapse
|
7
|
Immunotherapy and immunoengineering for breast cancer; a comprehensive insight into CAR-T cell therapy advancements, challenges and prospects. Cell Oncol (Dordr) 2022; 45:755-777. [PMID: 35943716 DOI: 10.1007/s13402-022-00700-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Breast cancer (BC) is a highly prevalent solid cancer with a high-rise infiltration of immune cells, turning it into a significant candidate for tumor-specific immunotherapies. Chimeric antigen receptor (CAR)-T cells are emerging as immunotherapeutic tools with genetically engineered receptors to efficiently recognize and attack tumor cells that express specific target antigens. Technological advancements in CAR design have provided five generations of CAR-T cells applicable to a wide range of cancer patients while boosting CAR-T cell therapy safety. However, CAR-T cell therapy is ineffective against breast cancer because of the loss of specified antigens, the immunosuppressive nature of the tumor and CAR-T cell-induced toxicities. Next-generation CAR-T cells actively pass through the tumor vascular barriers, persist for extended periods and disrupt the tumor microenvironment (TME) to block immune escape. CONCLUSION CAR-T cell therapy embodies advanced immunotherapy for BC, but further pre-clinical and clinical assessments are recommended to achieve maximized efficiency and safety.
Collapse
|
8
|
Wang L, Wang X, Guo E, Mao X, Miao S. Emerging roles of platelets in cancer biology and their potential as therapeutic targets. Front Oncol 2022; 12:939089. [PMID: 35936717 PMCID: PMC9355257 DOI: 10.3389/fonc.2022.939089] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/29/2022] [Indexed: 12/15/2022] Open
Abstract
The main role of platelets is to control bleeding and repair vascular damage via thrombosis. They have also been implicated to promote tumor metastasis through platelet-tumor cell interactions. Platelet-tumor cell interactions promote tumor cell survival and dissemination in blood circulation. Tumor cells are known to induce platelet activation and alter platelet RNA profiles. Liquid biopsies based on tumor-educated platelet biomarkers can detect tumors and correlate with prognosis, personalized therapy, treatment monitoring, and recurrence prediction. Platelet-based strategies for cancer prevention and tumor-targeted therapy include developing drugs that target platelet receptors, interfere with the release of platelet particles, inhibit platelet-specific enzymes, and utilize platelet-derived “nano-platelets” as a targeted drug delivery platform for tumor therapy. This review elaborates on platelet-tumor cell interactions and the molecular mechanisms and discusses future research directions for platelet-based liquid biopsy techniques and platelet-targeted anti-tumor strategies.
Collapse
Affiliation(s)
- Lei Wang
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xueying Wang
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Erliang Guo
- Department of Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xionghui Mao
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Xionghui Mao, ; Susheng Miao,
| | - Susheng Miao
- Department of Head and Neck Surgery, Harbin Medical University Cancer Hospital, Harbin, China
- *Correspondence: Xionghui Mao, ; Susheng Miao,
| |
Collapse
|
9
|
The posttraumatic response of CD4+ regulatory T cells is modulated by direct cell-cell contact via CD40L- and P-selectin-dependent pathways. Cent Eur J Immunol 2021; 46:283-294. [PMID: 34764800 PMCID: PMC8574106 DOI: 10.5114/ceji.2021.109171] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 05/14/2021] [Indexed: 12/14/2022] Open
Abstract
CD4+ FoxP3+ regulatory T cells (CD4+ Tregs) are important for the posttraumatic anti-inflammatory host response. As described previously, platelets are able to modulate CD4+ Treg activity in a reciprocally activating interaction following injury. The underlying mechanisms of the posttraumatic interaction between platelets and CD4+ Tregs remain unclear. We investigated the potential influence of CD40L and P-selectin, molecules known to be involved in direct cell contact of these cell types. In a murine burn injury model, the potential interaction pathways were addressed using CD40L- and P-selectin-deficient mice. Draining lymph nodes were harvested following trauma (1 h) and following a sham procedure. Early rapid activation of CD4+ Tregs was assessed by phospho-flow cytometry (signaling molecules (p)PKC-δ and (p)ZAP-70). Platelet function was analyzed performing rotational thromboelastometry (ROTEM). We hypothesized that disruption of the direct cell-cell contact via CD40L and P-selectin would affect posttraumatic activation of CD4+ Tregs and influence the hemostatic function of platelets. Indeed, while injury induced early activation of CD4+ Tregs in wild-type mice (ZAP-70: p = 0.13, pZAP-70: p < 0.05, PKC-δ: p < 0.05, pPKC-δ: p < 0.05), disruption of CD40L-dependent interaction (ZAP-70: p = 0.57, pZAP-70: p = 0.68, PKC-δ: p = 0.68, pPKC-δ: p = 0.9) or P-selectin-dependent interaction (ZAP-70: p = 0.78, pZAP-70: p = 0.58, PKC-δ: p = 0.81, pPKC-δ: p = 0.73) resulted in reduced posttraumatic activation. Furthermore, hemostatic function was impaired towards hypocoagulability in either deficiency. Our results suggest that the posttraumatic activation of CD4+ Tregs and hemostatic function of platelets are affected by direct cell-cell-signaling via CD40L and P-selectin.
Collapse
|
10
|
Liu Y, Zhang Y, Ding Y, Zhuang R. Platelet-mediated tumor metastasis mechanism and the role of cell adhesion molecules. Crit Rev Oncol Hematol 2021; 167:103502. [PMID: 34662726 DOI: 10.1016/j.critrevonc.2021.103502] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 09/17/2021] [Accepted: 10/10/2021] [Indexed: 12/12/2022] Open
Abstract
Mounting evidence suggests that platelets play an essential role in cancer metastasis. The interactions between platelets and circulating tumor cells (CTCs) promote cancer metastasis. CTCs induce platelet activation and aggregation, and activated platelets gather and protect CTCs from shear stress and natural killer cells. Finally, platelets stimulate CTC anoikis resistance, epithelial-to-mesenchymal transition, angiogenesis, extravasation, and eventually, metastasis. Cell adhesion molecules (CAMs) have been identified as active players during the interaction of CTCs with platelets, but the specific mechanism underlying the contribution of platelet-associated CAMs to CTC metastasis remains unclear. In this review, we introduce the mechanism of platelet-related tumor metastasis and particularly focus on the role of CAMs in it.
Collapse
Affiliation(s)
- Yitian Liu
- Department of Immunology, the Fourth Military Medical University, #169 Changlexilu Road, Xi'an, Shaanxi, 710032, China; Orthopedic Department of Tangdu Hospital, the Fourth Military Medical University, #1 Xinsi Road, Xi'an, Shaanxi, 710032, China
| | - Yuan Zhang
- Institute of Medical Research, Northwestern Polytechnical University, #127 Youyixilu Road, Xi'an, Shaanxi, 710072, China
| | - Yong Ding
- Orthopedic Department of Tangdu Hospital, the Fourth Military Medical University, #1 Xinsi Road, Xi'an, Shaanxi, 710032, China
| | - Ran Zhuang
- Department of Immunology, the Fourth Military Medical University, #169 Changlexilu Road, Xi'an, Shaanxi, 710032, China; Institute of Medical Research, Northwestern Polytechnical University, #127 Youyixilu Road, Xi'an, Shaanxi, 710072, China.
| |
Collapse
|
11
|
Feng Q, Wang M, Muhtar E, Wang Y, Zhu H. Nanoparticles of a New Small-Molecule P-Selectin Inhibitor Attenuate Thrombosis, Inflammation, and Tumor Growth in Two Animal Models. Int J Nanomedicine 2021; 16:5777-5795. [PMID: 34471352 PMCID: PMC8403725 DOI: 10.2147/ijn.s316863] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/18/2021] [Indexed: 12/26/2022] Open
Abstract
Purpose To assess whether the newly designed small-molecule oral P-selectin inhibitor 3S-1,2,3,4-tetrahydro-β-carboline-3-methyl aspartyl ester (THCMA) as a nanomedicine enhances antithrombosis, anti-inflammation, and antitumor activity more than the clinical trial drug PSI-697. Methods THCMA was designed as an amphiphile containing pharmacophores of PSI-697. Its nanofeatures were explored with TEM, SEM, Tyndall effect, ζ-potential, FT-ICR-MS, and NOESY 2D 1H NMR. The P-selectin inhibitory effect of THCMA was demonstrated with molecular docking, ultraviolet (UV) spectra, and competitive ELISA. In vivo and in vitro assays — anti-arterial thrombosis, anti–venous thrombosis, anti-inflammation, antitumor growth, anti–platelet aggregation, rat-tail bleeding time, anticoagulation index, soluble P-selectin (sP-selectin) expression, and serum TNFα expression — were performed to explore bioactivity and potential mechanisms. Water solubility of THCMA was measured using UV-absorption spectra. Results THCMA self-assembled into nanorings of approximately 100 nm in diameter. Its water solubility was about 1,030-fold that of PSI-697. THCMA exhibited more potent P-selectin inhibitory effect than PSI-697. The oral efficacy of THCMA was 100-fold that of PSI-697 in inhibiting arterial and venous thrombosis and tenfold in inhibiting inflammation. THCMA inhibited thrombosis at a dose that produces no coagulation disorders and no bleeding risk. THCMA exhibited enhanced antitumor activity over PSI-697 without systemic chemotherapy toxicity. THCMA significantly inhibited platelet aggregation in vitro and downregulated the expression levels of serum sP-selectin and TNFα in vivo. Conclusion A new small-molecule P-selectin inhibitor, THCMA, has been successfully designed as a nanomedicine with largely enhanced oral efficacy compared to the clinical trial drug PSI-697, and thus might be developed for the oral treatment of arterial thrombosis, venous thrombosis, inflammation, and cancer-associated thrombosis.
Collapse
Affiliation(s)
- Qiqi Feng
- School of Pharmaceutical Sciences, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Mengyang Wang
- School of Pharmaceutical Sciences, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Eldar Muhtar
- School of Pharmaceutical Sciences, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Yaonan Wang
- School of Pharmaceutical Sciences, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Capital Medical University, Beijing, 100069, People's Republic of China
| | - Haimei Zhu
- School of Pharmaceutical Sciences, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Capital Medical University, Beijing, 100069, People's Republic of China
| |
Collapse
|
12
|
DeRogatis JM, Viramontes KM, Neubert EN, Tinoco R. PSGL-1 Immune Checkpoint Inhibition for CD4 + T Cell Cancer Immunotherapy. Front Immunol 2021; 12:636238. [PMID: 33708224 PMCID: PMC7940186 DOI: 10.3389/fimmu.2021.636238] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/04/2021] [Indexed: 01/15/2023] Open
Abstract
Immune checkpoint inhibition targeting T cells has shown tremendous promise in the treatment of many cancer types and are now standard therapies for patients. While standard therapies have focused on PD-1 and CTLA-4 blockade, additional immune checkpoints have shown promise in promoting anti-tumor immunity. PSGL-1, primarily known for its role in cellular migration, has also been shown to function as a negative regulator of CD4+ T cells in numerous disease settings including cancer. PSGL-1 is highly expressed on T cells and can engage numerous ligands that impact signaling pathways, which may modulate CD4+ T cell differentiation and function. PSGL-1 engagement in the tumor microenvironment may promote CD4+ T cell exhaustion pathways that favor tumor growth. Here we highlight that blocking the PSGL-1 pathway on CD4+ T cells may represent a new cancer therapy approach to eradicate tumors.
Collapse
Affiliation(s)
| | | | | | - Roberto Tinoco
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
13
|
Mendoza-Almanza G, Burciaga-Hernández L, Maldonado V, Melendez-Zajgla J, Olmos J. Role of platelets and breast cancer stem cells in metastasis. World J Stem Cells 2020; 12:1237-1254. [PMID: 33312396 PMCID: PMC7705471 DOI: 10.4252/wjsc.v12.i11.1237] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/23/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023] Open
Abstract
The high mortality rate of breast cancer is mainly caused by the metastatic ability of cancer cells, resistance to chemotherapy and radiotherapy, and tumor regression capacity. In recent years, it has been shown that the presence of breast cancer stem cells is closely associated with the migration and metastatic ability of cancer cells, as well as with their resistance to chemotherapy and radiotherapy. The tumor microenvironment is one of the main molecular factors involved in cancer and metastatic processes development, in this sense it is interesting to study the role of platelets, one of the main communicator cells in the human body which are activated by the signals they receive from the microenvironment and can generate more than one response. Platelets can ingest and release RNA, proteins, cytokines and growth factors. After the platelets interact with the tumor microenvironment, they are called "tumor-educated platelets." Tumor-educated platelets transport material from the tumor microenvironment to sites adjacent to the tumor, thus helping to create microenvironments conducive for the development of primary and metastatic tumors. It has been observed that the clone capable of carrying out the metastatic process is a cancer cell with stem cell characteristics. Cancer stem cells go through a series of processes, including epithelial-mesenchymal transition, intravasation into blood vessels, movement through blood vessels, extravasation at the site of the establishment of a metastatic focus, and site colonization. Tumor-educated platelets support all these processes.
Collapse
Affiliation(s)
| | | | - Vilma Maldonado
- Laboratorio de Epigenética, Instituto Nacional de Medicina Genómica, Ciudad de México 14610, Mexico
| | - Jorge Melendez-Zajgla
- Génómica funcional del cáncer, Instituto Nacional de Medicina Genómica, Ciudad de México 14610, Mexico
| | - Jorge Olmos
- Biotecnología Marina, Centro de Investigación Científica y de Estudios Superiores de Ensenada, Ensenada 22860, Mexico
| |
Collapse
|
14
|
Li Z, Zheng Z, Li C, Li Z, Wu J, Zhang B. Therapeutic drugs and drug delivery systems targeting stromal cells for cancer therapy: a review. J Drug Target 2020; 28:714-726. [DOI: 10.1080/1061186x.2020.1744157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Zhaohuan Li
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Zengjuan Zheng
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Chenglei Li
- School of Pharmacy, Weifang Medical University, Weifang, China
| | - Zhipeng Li
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Jingliang Wu
- School of Bioscience and Technology, Weifang Medical University, Weifang, China
| | - Bo Zhang
- School of Pharmacy, Weifang Medical University, Weifang, China
| |
Collapse
|
15
|
Chen H, Lu A, Zhang X, Gui L, Wang Y, Wu J, Feng H, Peng S, Zhao M. Design and development of ICCA as a dual inhibitor of GPIIb/IIIa and P-selectin receptors. Drug Des Devel Ther 2018; 12:2097-2110. [PMID: 30022809 PMCID: PMC6042529 DOI: 10.2147/dddt.s169238] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The impact of upregulation of platelet membrane glycoprotein (GP)IIb/IIIa and P-selectin on the onset of arterial thrombosis, venous thrombosis, and cancer encourages to hypothesize that dual inhibitor of GPIIb/IIIa and P-selectin receptors should simultaneously inhibit arterial thrombosis, block venous thrombosis, and slow tumor growth. METHODS For this reason, the structural characteristics and the CDOCKER interaction energies of 12 carbolines were analyzed. This led to the design of 1-(4-isopropyl-phenyl)-β-carboline-3-carboxylic acid (ICCA) as a promising inhibitor of GPIIb/IIIa and P-selectin receptors. RESULTS The synthetic route provided ICCA in 48% total yield and 99.6% high-performance liquid chromatography purity. In vivo 5 μmol/kg oral ICCA downregulated GPIIb/IIIa and P-selectin expression thereby inhibited arterial thrombosis, blocked venous thrombosis, and slowed down tumor growth, but did not damage the kidney and the liver. CONCLUSION Therefore, ICCA could be a promising candidate capable of downregulating GPIIb/IIIa and P-selectin receptors, inhibiting arterial thrombosis, blocking venous thrombosis, and slowing down tumor growth.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/chemical synthesis
- Antibiotics, Antineoplastic/chemistry
- Antibiotics, Antineoplastic/pharmacology
- Carbolines/chemical synthesis
- Carbolines/chemistry
- Carbolines/pharmacology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Dose-Response Relationship, Drug
- Down-Regulation/drug effects
- Doxorubicin/chemical synthesis
- Doxorubicin/chemistry
- Doxorubicin/pharmacology
- Drug Design
- Drug Screening Assays, Antitumor
- Humans
- Male
- Mice
- Mice, Inbred ICR
- Models, Molecular
- Molecular Structure
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- P-Selectin/antagonists & inhibitors
- P-Selectin/metabolism
- Platelet Aggregation/drug effects
- Platelet Aggregation Inhibitors/chemical synthesis
- Platelet Aggregation Inhibitors/chemistry
- Platelet Aggregation Inhibitors/pharmacology
- Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors
- Platelet Glycoprotein GPIIb-IIIa Complex/metabolism
- Rats
- Rats, Sprague-Dawley
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Haiyan Chen
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
| | - An Lu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
| | - Xiaoyi Zhang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
| | - Lin Gui
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
| | - Yaonan Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
| | - Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
| | - Hua Feng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan, ;
| |
Collapse
|
16
|
Gareau AJ, Brien C, Gebremeskel S, Liwski RS, Johnston B, Bezuhly M. Ticagrelor inhibits platelet-tumor cell interactions and metastasis in human and murine breast cancer. Clin Exp Metastasis 2018; 35:25-35. [PMID: 29322294 DOI: 10.1007/s10585-018-9874-1] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 01/06/2018] [Indexed: 12/21/2022]
Abstract
Activated platelets promote the proliferation and metastatic potential of cancer cells. Platelet activation is largely mediated through ADP engagement of purinergic P2Y12 receptors on platelets. We examined the potential of the reversible P2Y12 inhibitor ticagrelor, an agent used clinically to prevent cardiovascular and cerebrovascular events, to reduce tumor growth and metastasis. In vitro, MCF-7, MDA-MB-468, and MDA-MB-231 human mammary carcinoma cells exhibited decreased interaction with platelets treated with ticagrelor compared to untreated platelets. Prevention of tumor cell-platelet interactions through pretreatment of platelets with ticagrelor did not improve natural killer cell-mediated tumor cell killing of K562 myelogenous leukemia target cells. Additionally, ticagrelor had no effect on proliferation of 4T1 mouse mammary carcinoma cells co-cultured with platelets, or on primary 4T1 tumor growth. In an orthotopic 4T1 breast cancer model, ticagrelor (10 mg/kg), but not clopidogrel (10 mg/kg) or saline, resulted in reduced metastasis and improved survival. Ticagrelor treatment was associated with a marked reduction in tumor cell-platelet aggregates in the lungs at 10, 30 and 60 min post-intravenous inoculation. These findings suggest a role for P2Y12-mediated platelet activation in promoting metastasis, and provide support for the use of ticagrelor in the prevention of breast cancer spread.
Collapse
Affiliation(s)
- Alison J Gareau
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada.,Izaak Walton Killam Health Centre, Halifax, NS, Canada
| | - Colin Brien
- School of Medicine, University College Cork, Cork, Ireland
| | - Simon Gebremeskel
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada
| | - Robert S Liwski
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada.,Department of Pathology, Dalhousie University, Halifax, NS, Canada
| | - Brent Johnston
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada.,Department of Pathology, Dalhousie University, Halifax, NS, Canada.,Department of Pediatrics, Dalhousie University, Halifax, NS, Canada
| | - Michael Bezuhly
- Department of Microbiology & Immunology, Dalhousie University, Halifax, NS, Canada. .,Izaak Walton Killam Health Centre, Halifax, NS, Canada. .,Division of Plastic and Reconstructive Surgery, Department of Surgery, Dalhousie University, Halifax, NS, Canada. .,Department of Surgery, Dalhousie University, IWK Health Centre, 5850/5980 University Avenue, PO Box 9700, Halifax, NS, B3K 6R8, Canada.
| |
Collapse
|
17
|
Zhu H, Wang Y, Song C, Feng Q, Wu J, Zhao S, Gui L, Zhang X, Zhao M, Peng S. Docking of THPDTPI: to explore P-selectin as a common target of anti-tumor, anti-thrombotic and anti-inflammatory agent. Oncotarget 2018; 9:268-281. [PMID: 29416612 PMCID: PMC5787463 DOI: 10.18632/oncotarget.19374] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 07/06/2017] [Indexed: 01/01/2023] Open
Abstract
The impact of soluble P-selectin on tumor growth, thrombosis and inflammation has been individually documented. Whether the down-regulation of P-selectin expression can simultaneously slow the tumor growth, inhibit the thrombosis and attenuate the inflammatory response remains unknown. In this context, (2'S,5'S)- tetrahydropyrazino[1',2':1,6]-di{2,3,4,9-tetrahydro-1H-pyrido[3,4-b]indole}-1',4'-dione (THPDTPI) was designed as an inhibitor of P-selectin. The suitable docking of THPDTPI towards the active site of P-selectin, the significant down-regulation of THPDTPI to P-selectin expression, and the direct action of THPDTPI on P-selectin suggest that P-selectin could be a target of THPDTPI. In vivo THPDTPI possesses the anti-tumor activity, the anti-thrombotic activity and the anti-inflammatory activity. This implies that targeting P-selectin is of essential importance for this triple activity. The minimal effective doses of THPDTPI inhibiting the tumor growth, the rat arterial thrombosis and the mouse ear edema are 0.01 μmol/kg, 0.1 μmol/kg and 0.001 μmol/kg, respectively. Atomic force microscopy images and FT-MS spectra showed that the adhesion of THPDTPI onto the surfaces of the platelets may be the first step of P-selectin targeting. Besides, the dependence of the triple action of THPDTPI inhibiting the tumor growth, the thrombosis and the inflammation on the decrease of the soluble P-selectin led to the correlation of the soluble P-selectin with the serum TNF-α and serum IL-8.
Collapse
Affiliation(s)
- Haimei Zhu
- College of Pharmaceutical Sciences of Capital Medical University, Beijing, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing, China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
| | - Yuji Wang
- College of Pharmaceutical Sciences of Capital Medical University, Beijing, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing, China
- Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Ce Song
- Guangxi Pusen Biotechnology Co. Ltd., Guilin, China
| | - Qiqi Feng
- College of Pharmaceutical Sciences of Capital Medical University, Beijing, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing, China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
| | - Jianhui Wu
- College of Pharmaceutical Sciences of Capital Medical University, Beijing, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing, China
- Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Shurui Zhao
- College of Pharmaceutical Sciences of Capital Medical University, Beijing, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing, China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
| | - Lin Gui
- College of Pharmaceutical Sciences of Capital Medical University, Beijing, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing, China
- Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Xiaoyi Zhang
- College of Pharmaceutical Sciences of Capital Medical University, Beijing, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing, China
- Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Ming Zhao
- College of Pharmaceutical Sciences of Capital Medical University, Beijing, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing, China
- Beijing Laboratory of Biomedical Materials, Beijing, China
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shiqi Peng
- College of Pharmaceutical Sciences of Capital Medical University, Beijing, China
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing, China
- Beijing Laboratory of Biomedical Materials, Beijing, China
| |
Collapse
|
18
|
The Interaction of Selectins and PSGL-1 as a Key Component in Thrombus Formation and Cancer Progression. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6138145. [PMID: 28680883 PMCID: PMC5478826 DOI: 10.1155/2017/6138145] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 04/12/2017] [Accepted: 04/23/2017] [Indexed: 12/11/2022]
Abstract
Cellular interaction is inevitable in the pathomechanism of human disease. Formation of heterotypic cellular aggregates, between distinct cells of hematopoietic and nonhematopoietic origin, may be involved in events leading to inflammation and the complex process of cancer progression. Among adhesion receptors, the family of selectins with their ligands have been considered as one of the major contributors to cell-cell interactions. Consequently, the inhibition of the interplay between selectins and their ligands may have potential therapeutic benefits. In this review, we focus on the current evidence on the selectins as crucial modulators of inflammatory, thrombotic, and malignant disorders. Knowing that there is promiscuity in selectin binding, we outline the importance of a key protein that serves as a ligand for all selectins. This dimeric mucin, the P-selectin glycoprotein ligand 1 (PSGL-1), has emerged as a major player in inflammation, thrombus, and cancer development. We discuss the interaction of PSGL-1 with various selectins in physiological and pathological processes with particular emphasis on mechanisms that lead to severe disease.
Collapse
|
19
|
Gregório AC, Fonseca NA, Moura V, Lacerda M, Figueiredo P, Simões S, Dias S, Moreira JN. Inoculated Cell Density as a Determinant Factor of the Growth Dynamics and Metastatic Efficiency of a Breast Cancer Murine Model. PLoS One 2016; 11:e0165817. [PMID: 27820870 PMCID: PMC5098815 DOI: 10.1371/journal.pone.0165817] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 10/18/2016] [Indexed: 01/16/2023] Open
Abstract
4T1 metastatic breast cancer model have been widely used to study stage IV human breast cancer. However, the frequent inoculation of a large number of cells, gives rise to fast growing tumors, as well as to a surprisingly low metastatic take rate. The present work aimed at establishing the conditions enabling high metastatic take rate of the triple-negative murine 4T1 syngeneic breast cancer model. An 87% 4T1 tumor incidence was observed when as few as 500 cancer cells were implanted. 4T1 cancer cells colonized primarily the lungs with 100% efficiency, and distant lesions were also commonly identified in the mesentery and pancreas. The drastic reduction of the number of inoculated cells resulted in increased tumor doubling times and decreased specific growth rates, following a Gompertzian tumor expansion. The established conditions for the 4T1 mouse model were further validated in a therapeutic study with peguilated liposomal doxorubicin, in clinical used in the setting of metastatic breast cancer. Inoculated cell density was proven to be a key methodological aspect towards the reproducible development of macrometastases in the 4T1 mouse model and a more reliable pre-clinical assessment of antimetastatic therapies.
Collapse
Affiliation(s)
- Ana C. Gregório
- CNC—Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- IIIUC–Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Nuno A. Fonseca
- CNC—Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- FFUC—Faculty of Pharmacy, Pólo das Ciências da Saúde, University of Coimbra, Coimbra, Portugal
| | - Vera Moura
- CNC—Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- TREAT U, SA, Coimbra, Portugal
| | - Manuela Lacerda
- IPATIMUP–Institute of Molecular Pathology and Immunology, University of Porto, Porto, Portugal
| | - Paulo Figueiredo
- IPOFG-EPE–Portuguese Institute of Oncology Francisco Gentil, Coimbra, Portugal
| | - Sérgio Simões
- CNC—Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- FFUC—Faculty of Pharmacy, Pólo das Ciências da Saúde, University of Coimbra, Coimbra, Portugal
| | - Sérgio Dias
- IMM–Institute of Molecular Medicine, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| | - João Nuno Moreira
- CNC—Center for Neurosciences and Cell Biology, University of Coimbra, Coimbra, Portugal
- FFUC—Faculty of Pharmacy, Pólo das Ciências da Saúde, University of Coimbra, Coimbra, Portugal
- * E-mail:
| |
Collapse
|
20
|
Xu W, Zhao M, Wang Y, Zhu H, Wang Y, Zhao S, Wu J, Peng S. Design, synthesis, and in vivo evaluations of benzyl N ω-nitro-N α-(9H-pyrido[3,4-b]indole-3-carbonyl)- l-argininate as an apoptosis inducer capable of decreasing the serum concentration of P-selectin. MEDCHEMCOMM 2016. [DOI: 10.1039/c6md00215c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
A series of findings suggest that the discovery of in vivo apoptosis inducers for chemotherapy is of clinical importance.
Collapse
Affiliation(s)
- Wenyun Xu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences
- Capital Medical University
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences
- Capital Medical University
| | - Yuji Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences
- Capital Medical University
| | - Haimei Zhu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences
- Capital Medical University
| | - Yaonan Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences
- Capital Medical University
| | - Shurui Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences
- Capital Medical University
| | - Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences
- Capital Medical University
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Beijing Laboratory of Biomedical Materials
- College of Pharmaceutical Sciences
- Capital Medical University
| |
Collapse
|