1
|
Yegin ZA, Yıldız Ş, Savaş EM, Şeker A, Uyar Göçün P, Özkurt ZN, Akyürek N, Yağcı M. Day + 100 bone marrow megakaryocyte count predicts transplant outcome in patients with high-risk myelodysplastic syndrome and acute leukemia. J Hematop 2025; 18:18. [PMID: 40232552 DOI: 10.1007/s12308-025-00633-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/05/2025] [Indexed: 04/16/2025] Open
Abstract
Megakaryocytes (MKs), the precursor cells of platelets, have essential roles in a variety of pathophysiological processes in the bone marrow (BM) niche. Megakaryocytes maintain hematopoietic stem cell microenvironment through inflammatory and immunological responses. The primary objective of this research was to investigate the clinical impact of BM-MK counts in high-risk myelodysplastic syndrome and acute leukemia patients who underwent allogeneic hematopoietic stem cell transplantation (alloHSCT). Three hundred and forty-six patients (median age, 42 (15-71) years; male/female, 207/139) participated in the study. Based on the BM-MK counts on day + 100 of alloHSCT, the study population was classified into normal/high-MK+100 and low-MK+100 groups. The probabilities of progression-free survival (PFS) and overall survival (OS) were significantly better in the normal/high-MK+100 group (p < 0.001, p < 0.001). Nonrelapse mortality was found to be reduced in the same group of patients (p = 0.012). BM-MK+100 count, which was indicated to be a predictor for relapse after alloHSCT (p = 0.018), represented a considerable impact on PFS and OS (p = 0.017, p = 0.009). Megakaryocytes have regulatory roles in association with a comprehensive cytokine network in the BM microenvironment. Although the localization of MKs may be determinative for their spectrum of efficacy, distinct biological subgroups may also help to clarify the heterogeneity of their functional features. Prospective efforts in larger populations are required to illuminate the potential prognostic role of MKs in alloHSCT recipients.
Collapse
|
2
|
Xu B, Ye X, Sun K, Chen L, Wen Z, Lan Q, Chen J, Chen M, Shen M, Wang S, Xu Y, Zhang X, Zhao J, Wang J, Chen S. IRAP Drives Ribosomal Degradation to Refuel Energy for Platelet Activation during Septic Thrombosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411914. [PMID: 39853919 PMCID: PMC11967848 DOI: 10.1002/advs.202411914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/09/2025] [Indexed: 01/26/2025]
Abstract
Platelets play crucial roles in multiple pathophysiological processes after energy-dependent activation. It is puzzling how such a small cellular debris has abundant energy supply. In this study, it is shown that insulin-regulated aminopeptidase (IRAP), a type II transmembrane protein, is a key regulator for platelet activation by promoting energy regeneration during septic thrombosis. Through interaction with certain endosome membrane proteins, IRAP can not only promote granule release, but also facilitate lysosomal degradation of theoretically discarded ribosomes in an mTORC1- and S-acylation-dependent manner in activated platelets. Plentiful amino acids obtained from IRAP-mediated ribophagy are recruited to aerobic glycolysis and then promote energy metabolism reprogramming, thereby producing abundant energy for platelet life extension and prolonged activation. Consequently, targeted blocking IRAP can dramatically alleviate platelet hyperactivation and septic thrombosis.
Collapse
Affiliation(s)
- Baichuan Xu
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Xianpeng Ye
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Kangfu Sun
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Liang Chen
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Zhaoyang Wen
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Qigang Lan
- Department of NephrologyChongqing Key Laboratory of Prevention and Treatment of Kidney DiseaseChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Jun Chen
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Mo Chen
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Mingqiang Shen
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Song Wang
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Yang Xu
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Xi Zhang
- Medical Center of HematologyXinqiao HospitalState Key Laboratory of Trauma and Chemical PoisoningArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Jinghong Zhao
- Department of NephrologyChongqing Key Laboratory of Prevention and Treatment of Kidney DiseaseChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Junping Wang
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Shilei Chen
- State Key Laboratory of Trauma and Chemical PoisoningInstitute of Combined InjuryChongqing Engineering Research Center for NanomedicineCollege of Preventive MedicineArmy Medical University (Third Military Medical University)Chongqing400038China
| |
Collapse
|
3
|
Josefsson EC. Platelets and megakaryocytes in cancer. J Thromb Haemost 2025; 23:804-816. [PMID: 39742972 DOI: 10.1016/j.jtha.2024.12.016] [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: 10/06/2024] [Revised: 12/03/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
Platelets have important roles in hemostasis but also actively participate in cancer metastasis and inflammatory processes. They are produced by large precursor cells, the megakaryocytes, residing mainly in the bone marrow. Clinically, elevated platelet counts and/or increased platelet-to-lymphocyte ratio are being explored as biomarkers of metastatic disease and to predict survival or response to therapy in certain cancers. Multiple mechanisms have been put forward on how platelets promote hematogenous metastasis stemming mainly from murine experimental models. Research is now beginning to explore the potential roles of megakaryocytes in solid cancer, myeloma, and lymphoma. Here, we review mechanisms on how platelets and megakaryocytes contribute to cancer progression and metastasis but also discuss potential cancer-suppressing functions mainly related to the regulation of vascular intratumor integrity. Recent developments in cancer immune checkpoint therapy are reviewed with a focus on the potential roles of platelets. Moreover, we review studies exploring platelets for targeted drug delivery systems in cancer therapy.
Collapse
Affiliation(s)
- Emma C Josefsson
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Chemistry, Gothenburg, Sweden; Department of Laboratory Medicine, Institute of Biomedicine, The University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
4
|
Hu X, He Y, Li S, Jiang Y, Yu R, Wu Y, Fu X, Song Y, Lin C, Shi J, Li HB, Gao Y. Acute inflammation induces acute megakaryopoiesis with impaired platelet production during fetal hematopoiesis. Development 2025; 152:dev204226. [PMID: 39817838 DOI: 10.1242/dev.204226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 12/27/2024] [Indexed: 01/18/2025]
Abstract
Hematopoietic development is tightly regulated by various factors. The role of RNA m6A modification during fetal hematopoiesis, particularly in megakaryopoiesis, remains unclear. Here, we demonstrate that loss of m6A methyltransferase METTL3 induces formation of double-stranded RNAs (dsRNAs) and activates acute inflammation during fetal hematopoiesis in mouse. This dsRNA-mediated inflammation leads to acute megakaryopoiesis, which facilitates the generation of megakaryocyte progenitors but disrupts megakaryocyte maturation and platelet production. The inflammation and immune response activate the phosphorylation of STAT1 and IRF3, and upregulate downstream interferon-stimulated genes (ISGs). Inflammation inhibits the proliferation rate of hematopoietic progenitors and further skews the cell fate determination toward megakaryocytes rather than toward erythroid from megakaryocyte-erythroid progenitors (MEPs). Transcriptional-wide gene expression analysis identifies IGF1 as a major factor whose reduction is responsible for the inhibition of megakaryopoiesis and thrombopoiesis. Restoration of IGF1 with METTL3-deficient hematopoietic cells significantly increases megakaryocyte maturation. In summary, we elucidate that the loss of RNA m6A modification-induced acute inflammation activates acute megakaryopoiesis, but impairs its final maturation through the inhibition of IGF1 expression during fetal hematopoiesis.
Collapse
Affiliation(s)
- Xiaojie Hu
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yirui He
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Shengwei Li
- Department of General Surgery, Shanghai Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yue Jiang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Renjie Yu
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yi Wu
- Fundamental Research Center, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Xiaoying Fu
- Department of Laboratory Medicine, Shenzhen Children's Hospital, Shenzhen, Guangdong 518000, China
| | - Yuanbin Song
- Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou 510006, 510006 Guangdong, China
| | - Changdong Lin
- Fundamental Research Center, Shanghai YangZhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China
| | - Jiejun Shi
- Department of General Surgery, Shanghai Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Hua-Bing Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Institute of Immunological Innovation & Translation, Chongqing Medical University, Chongqing 400016, China
| | - Yimeng Gao
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai 200092, China
| |
Collapse
|
5
|
Hu L, Zheng C, Kong Y, Luo Z, Huang F, Zhu Z, Li Q, Liang M. Cathepsin G promotes arteriovenous fistula maturation by positively regulating the MMP2/MMP9 pathway. Ren Fail 2024; 46:2316269. [PMID: 38362707 PMCID: PMC10878333 DOI: 10.1080/0886022x.2024.2316269] [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: 10/18/2023] [Accepted: 02/04/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Arteriovenous fistula (AVF) is currently the preferred vascular access for hemodialysis patients. However, the low maturation rate of AVF severely affects its use in patients. A more comprehensive understanding and study of the mechanisms of AVF maturation is urgently needed. METHODS AND RESULTS In this study, we downloaded the publicly available datasets (GSE119296 and GSE220796) from the Gene Expression Omnibus (GEO) and merged them for subsequent analysis. We screened 84 differentially expressed genes (DEGs) and performed the functional enrichment analysis. Next, we integrated the results obtained from the degree algorithm provided by the Cytohubba plug-in, Molecular complex detection (MCODE) plug-in, weighted gene correlation network analysis (WGCNA), and Least absolute shrinkage and selection operator (LASSO) logistic regression. This integration allowed us to identify CTSG as a hub gene associated with AVF maturation. Through the literature search and Pearson's correlation analysis, the genes matrix metalloproteinase 2 (MMP2) and MMP9 were identified as potential downstream effectors of CTSG. We then collected three immature clinical AVF vein samples and three mature samples and validated the expression of CTSG using immunohistochemistry (IHC) and double-immunofluorescence staining. The IHC results demonstrated a significant decrease in CTSG expression levels in the immature AVF vein samples compared to the mature samples. The results of double-immunofluorescence staining revealed that CTSG was expressed in both the intima and media of AVF veins. Moreover, the expression of CTSG in vascular smooth muscle cells (VSMCs) was significantly higher in the mature samples compared to the immature samples. The results of Masson's trichrome and collagen I IHC staining demonstrated a higher extent of collagen deposition in the media of immature AVF veins compared to the mature. By constructing an in vitro CTSG overexpression model in VSMCs, we found that CTSG upregulated the expression of MMP2 and MMP9 while downregulating the expression of collagen I and collagen III. Furthermore, CTSG was found to inhibit VSMC migration. CONCLUSIONS CTSG may promote AVF maturation by stimulating the secretion of MMP2 and MMP9 from VSMCs and reducing the extent of medial fibrosis in AVF veins by inhibiting the secretion of collagen I and collagen III.
Collapse
Affiliation(s)
- Lemei Hu
- Department of Nephrology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, PRChina
- School of Medicine, South China University of Technology, Guangzhou, PRChina
| | - Changqing Zheng
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, PRChina
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, PR China
| | - Ying Kong
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, PRChina
| | - Zhiqing Luo
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, PRChina
| | - Fengzhang Huang
- Department of Nephrology, Guangzhou First People’s Hospital, Guangzhou, PRChina
| | - Zhigang Zhu
- Department of Geriatrics, Division of Hematology and Oncology, Second Affiliated Hospital, Guangzhou First People’s Hospital, College of Medicine, South China University of Technology, Guangzhou, PRChina
| | - Quhuan Li
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, PR China
- Guangdong Provincial Engineering and Technology Research Center of Biopharmaceuticals, South China University of Technology, Guangzhou, PR China
| | - Ming Liang
- Department of Nephrology, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, PRChina
- Department of Nephrology, Guangzhou First People’s Hospital, Guangzhou, PRChina
| |
Collapse
|
6
|
Wang Q, Chen X, Huang K, Deng G, Tian Y, Jiang K. S100A9 promotes renal calcium oxalate stone formation via activating the TLR4-p38/MAPK-LCN2 signaling pathway. Int J Biol Macromol 2024; 281:136178. [PMID: 39357728 DOI: 10.1016/j.ijbiomac.2024.136178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 08/20/2024] [Accepted: 09/29/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVES To explore the role of S100A9 protein in renal calcium oxalate (CaOx) stone formation. METHODS CaOx nephrocalcinosis mice were established via intraperitoneal injection of glyoxylate. They were treated with S100A9 deficiency, Paquinimod, or p38 MAPK-IN-1. Vonkossa staining was conducted to observe the deposition of CaOx crystals. Renal expression of inflammation, macrophage polarization, and injury markers was detected using immunohistochemistry and qPCR. Effects of S100A9 on renal tubular epithelial cells (HK-2) were explored by transcriptome sequencing. The mechanism of how S100A9 regulated lipocalin 2 (LCN2) was studied through Western Blot. Flow cytometry was performed to detect the influence of LCN2 on macrophages polarization. RESULTS S100A9 deficiency inhibited the renal deposition of CaOx crystals in nephrocalcinosis mice. S100A9 upregulated the expression of LCN2 in HK-2 cells via activating the TLR4-p38/MAPK pathway. LCN2 promoted the migration and M1 polarization of macrophages. S100A9 deficiency downregulated the renal expression of LCN2, IL1-β, Kim-1, F4/80, and CD80 in nephrocalcinosis mice. Paquinimod and p38 MAPK-IN-1 both inhibited the renal deposition of CaOx crystals and downregulated the expression of LCN2, IL1-β, Kim-1, F4/80, iNOS, and CD68 in nephrocalcinosis mice. CONCLUSIONS S100A9 promotes renal inflammatory injury by activating the TLR4-p38/MAPK-LCN2 pathway and then contributes to CaOx stone formation.
Collapse
Affiliation(s)
- Qing Wang
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550000, China.
| | - Xiaolong Chen
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550000, China
| | - Kunyuan Huang
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550000, China
| | - Guanyun Deng
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550000, China
| | - Yuan Tian
- Department of Urology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550000, China.
| | - Kehua Jiang
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550000, China.
| |
Collapse
|
7
|
Roweth HG, Becker IC, Malloy MW, Clarke EM, Munn SA, Kumar PL, Aivasovsky I, Tray K, Schmaier AA, Battinelli EM. Platelet Angiopoietin-1 Protects Against Murine Models of Tumor Metastasis. Arterioscler Thromb Vasc Biol 2024; 44:2024-2037. [PMID: 39051116 PMCID: PMC11335083 DOI: 10.1161/atvbaha.124.321189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND In addition to their fundamental roles in preserving vascular integrity, platelets also contribute to tumor angiogenesis and metastasis. However, despite being a reservoir for angiogenic and metastatic cytokines, platelets also harbor negative regulators of tumor progression. Angpt1 (angiopoietin-1) is a cytokine essential for developmental angiogenesis that also protects against tumor cell metastasis through an undefined mechanism. Although activated platelets release Angpt1 from α-granules into circulation, the contributions of platelet Angpt1 to tumor growth, angiogenesis, and metastasis have not been investigated. METHODS Using cytokine arrays and ELISAs, we first compared platelet Angpt1 levels in breast and melanoma mouse tumor models to tumor-free controls. We then assessed tumor growth and metastasis in mice lacking megakaryocyte and platelet Angpt1 (Angpt1Plt KO). The spontaneous metastasis of mammary-injected tumor cells to the lungs was quantified using RT-PCR (reverse transcription-polymerase chain reaction). The lung colonization of intravenously injected tumor cells and tumor cell extravasation were determined using fluorescent microscopy and flow cytometry. RESULTS Platelet Angpt1 is selectively upregulated in the PyMT (polyoma middle tumor antigen) breast cancer mouse model, and platelets are the principal source of Angpt1 in blood circulation. While primary tumor growth and angiogenesis were unaffected, Angpt1Plt KO mice had both increased spontaneous lung metastasis and tumor cell lung colonization following mammary or intravenous injection, respectively. Although platelet Angpt1 did not affect initial tumor cell entrapment in the lungs, Angpt1Plt KO mice had increased tumor cell retention and extravasation. Serum from Angpt1Plt KO mice increased endothelial permeability and reduced VE (vascular endothelial)-cadherin expression at endothelial junctions compared with serum from control mice (Angpt1WT). CONCLUSIONS Platelets provide an intravascular source of Angpt1 that restrains tumor metastasis by preserving the lung microvasculature to limit tumor cell extravasation.
Collapse
MESH Headings
- Animals
- Angiopoietin-1/genetics
- Angiopoietin-1/metabolism
- Angiopoietin-1/blood
- Blood Platelets/metabolism
- Blood Platelets/pathology
- Female
- Lung Neoplasms/secondary
- Lung Neoplasms/pathology
- Lung Neoplasms/genetics
- Lung Neoplasms/blood
- Lung Neoplasms/metabolism
- Lung Neoplasms/prevention & control
- Mice, Knockout
- Neovascularization, Pathologic
- Mice, Inbred C57BL
- Melanoma, Experimental/pathology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/blood
- Melanoma, Experimental/secondary
- Melanoma, Experimental/genetics
- Cell Line, Tumor
- Mice
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/blood
- Tumor Burden
- Disease Models, Animal
Collapse
Affiliation(s)
- Harvey G. Roweth
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA (H.G.R., M.W.M., E.M.C., S.A.M., P.L.K., E.M.B.)
- Harvard Medical School, Boston, MA (H.G.R., I.C.B., P.L.K., I.A., A.A.S., E.M.B.)
| | - Isabelle C. Becker
- Harvard Medical School, Boston, MA (H.G.R., I.C.B., P.L.K., I.A., A.A.S., E.M.B.)
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, MA (I.C.B.)
| | - Michael W. Malloy
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA (H.G.R., M.W.M., E.M.C., S.A.M., P.L.K., E.M.B.)
| | - Emily M. Clarke
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA (H.G.R., M.W.M., E.M.C., S.A.M., P.L.K., E.M.B.)
| | - Sophie A. Munn
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA (H.G.R., M.W.M., E.M.C., S.A.M., P.L.K., E.M.B.)
| | - Priya L. Kumar
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA (H.G.R., M.W.M., E.M.C., S.A.M., P.L.K., E.M.B.)
- Harvard Medical School, Boston, MA (H.G.R., I.C.B., P.L.K., I.A., A.A.S., E.M.B.)
| | - Ivan Aivasovsky
- Harvard Medical School, Boston, MA (H.G.R., I.C.B., P.L.K., I.A., A.A.S., E.M.B.)
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (I.A., K.T., A.A.S.)
| | - Kobe Tray
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (I.A., K.T., A.A.S.)
| | - Alec A. Schmaier
- Harvard Medical School, Boston, MA (H.G.R., I.C.B., P.L.K., I.A., A.A.S., E.M.B.)
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA (I.A., K.T., A.A.S.)
| | - Elisabeth M. Battinelli
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA (H.G.R., M.W.M., E.M.C., S.A.M., P.L.K., E.M.B.)
- Harvard Medical School, Boston, MA (H.G.R., I.C.B., P.L.K., I.A., A.A.S., E.M.B.)
| |
Collapse
|
8
|
Schaubaecher JB, Smiljanov B, Haring F, Steiger K, Wu Z, Ugurluoglu A, Luft J, Ballke S, Mahameed S, Schneewind V, Hildinger J, Canis M, Mittmann LA, Braun C, Zuchtriegel G, Kaiser R, Nicolai L, Mack M, Weichert W, Lauber K, Uhl B, Reichel CA. Procoagulant platelets promote immune evasion in triple-negative breast cancer. Blood 2024; 144:216-226. [PMID: 38648571 DOI: 10.1182/blood.2023022928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024] Open
Abstract
ABSTRACT Triple-negative breast cancer (TNBC) is an aggressive tumor entity in which immune checkpoint (IC) molecules are primarily synthesized in the tumor environment. Here, we report that procoagulant platelets bear large amounts of such immunomodulatory factors and that the presence of these cellular blood components in TNBC relates to protumorigenic immune-cell activity and impaired survival. Mechanistically, tumor-released nucleic acids attract platelets to the aberrant tumor microvasculature, where they undergo procoagulant activation, thus delivering specific stimulatory and inhibitory IC molecules. This concomitantly promotes protumorigenic myeloid leukocyte responses and compromises antitumorigenic lymphocyte activity, ultimately supporting tumor growth. Interference with platelet-leukocyte interactions prevented immune cell misguidance and suppressed tumor progression, nearly as effective as systemic IC inhibition. Hence, our data uncover a self-sustaining mechanism of TNBC by using platelets to misdirect immune-cell responses. Targeting this irregular multicellular interplay may represent a novel immunotherapeutic strategy for TNBC without the adverse effects of systemic IC inhibition.
Collapse
Affiliation(s)
- Johanna B Schaubaecher
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Bojan Smiljanov
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Florian Haring
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Katja Steiger
- Department of Pathology, Technical University Munich, Munich, Germany
| | - Zhengquan Wu
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Anais Ugurluoglu
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Joshua Luft
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Simone Ballke
- Department of Pathology, Technical University Munich, Munich, Germany
| | - Shaan Mahameed
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Vera Schneewind
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Jonas Hildinger
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Martin Canis
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Comprehensive Cancer Center, Munich Ludwig-Maximilians-Universität, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Laura A Mittmann
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Constanze Braun
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Gabriele Zuchtriegel
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Rainer Kaiser
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Medicine I, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Berlin, Germany
| | - Leo Nicolai
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Medicine I, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Berlin, Germany
| | - Matthias Mack
- Department of Nephrology, University of Regensburg, Regensburg, Germany
| | - Wilko Weichert
- Department of Pathology, Technical University Munich, Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Bernd Uhl
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| | - Christoph A Reichel
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Department of Otorhinolaryngology, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
- Comprehensive Cancer Center, Munich Ludwig-Maximilians-Universität, Ludwig-Maximilians-Universität University Hospital, Munich, Germany
| |
Collapse
|
9
|
Chen S, Looney MR. Understanding megakaryocyte phenotypes and the impact on platelet biogenesis. Transfusion 2024; 64:1372-1380. [PMID: 38923572 PMCID: PMC11251837 DOI: 10.1111/trf.17927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 06/28/2024]
Affiliation(s)
- Shiyu Chen
- Departments of Medicine and Laboratory Medicine, University
of California, San Francisco, San Francisco, CA, U.SA
| | - Mark R. Looney
- Departments of Medicine and Laboratory Medicine, University
of California, San Francisco, San Francisco, CA, U.SA
| |
Collapse
|
10
|
Bai L, Courcoubetis G, Mason J, Hicks JB, Nieva J, Kuhn P, Shishido SN. Longitudinal tracking of circulating rare events in the liquid biopsy of stage III-IV non-small cell lung cancer patients. Discov Oncol 2024; 15:142. [PMID: 38700626 PMCID: PMC11068717 DOI: 10.1007/s12672-024-00984-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/17/2024] [Indexed: 05/06/2024] Open
Abstract
In the United States, lung cancer is the second most common type of cancer with non-small cell lung cancer (NSCLC) encompassing around 85% of total lung cancer cases. Late-stage patients with metastatic disease have worsening prognosis, highlighting the importance of longitudinal disease monitoring. Liquid biopsy (LBx) represents a way for physicians to non-invasively track tumor analytes, such as circulating tumor cells (CTCs), and understand tumor progression in real-time through analyzing longitudinal blood samples. CTCs have been shown to be effective predictive biomarkers in measuring treatment efficacy and survival outcomes. We used the third-generation High-Definition Single Cell Assay (HDSCA3.0) workflow to analyze circulating rare events longitudinally during treatment in a cohort of 10 late-stage NSCLC patients, identifying rare events including circulating cancer cells (i.e., CTCs), and oncosomes. Here, we show (1) that there is a cancer specific LBx profile, (2) there is considerable heterogeneity of rare cells and oncosomes, and (3) that LBx data elements correlated with patient survival outcomes. Additional studies are warranted to understand the biological significance of the rare events detected, and the clinical potential of the LBx to monitor and predict response to treatment in NSCLC patient care.
Collapse
Affiliation(s)
- Lily Bai
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089, USA
| | - George Courcoubetis
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, 90089, USA
| | - Jeremy Mason
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, 90089, USA
- Catherine and Joseph Aresty Department of Urology, Institute of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - James B Hicks
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089, USA
| | - Jorge Nieva
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Peter Kuhn
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, 90089, USA.
- Department of Biological Sciences, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089, USA.
- Catherine and Joseph Aresty Department of Urology, Institute of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA.
| | - Stephanie N Shishido
- Convergent Science Institute in Cancer, Michelson Center for Convergent Bioscience, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
11
|
Abstract
Alongside their conventional roles in thrombosis and hemostasis, platelets have long been associated with nonhemostatic pathologies, including tumor cell metastasis. Numerous mechanistic studies have since demonstrated that the direct binding of platelets to intravascular tumor cells promotes key hallmarks of metastasis, including survival in circulation and tumor cell arrest at secondary sites. However, platelets also interact with nonmalignant cells that make up the stromal and immune compartments within both primary and metastatic tumors. This review will first provide a brief historical perspective on platelet contributions to metastatic disease before discussing the emerging roles that platelets play in creating microenvironments that likely support successful tumor cell metastasis.
Collapse
Affiliation(s)
- Harvey G. Roweth
- Hematology Division, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
12
|
Trivanović D, Mojsilović S, Bogosavljević N, Jurišić V, Jauković A. Revealing profile of cancer-educated platelets and their factors to foster immunotherapy development. Transl Oncol 2024; 40:101871. [PMID: 38134841 PMCID: PMC10776659 DOI: 10.1016/j.tranon.2023.101871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 12/03/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
Among multiple hemostasis components, platelets hyperactivity plays major roles in cancer progression by providing surface and internal components for intercellular crosstalk as well as by behaving like immune cells. Since platelets participate and regulate immunity in homeostatic and disease states, we assumed that revealing platelets profile might help in conceiving novel anti-cancer immune-based strategies. The goal of this review is to compile and discuss the most recent reports on the nature of cancer-associated platelets and their interference with immunotherapy. An increasing number of studies have emphasized active communication between cancer cells and platelets, with platelets promoting cancer cell survival, growth, and metastasis. The anti-cancer potential of platelet-directed therapy has been intensively investigated, and anti-platelet agents may prevent cancer progression and improve the survival of cancer patients. Platelets can (i) reduce antitumor activity; (ii) support immunoregulatory cells and factors generation; (iii) underpin metastasis and, (iv) interfere with immunotherapy by expressing ligands of immune checkpoint receptors. Mediators produced by tumor cell-induced platelet activation support vein thrombosis, constrain anti-tumor T- and natural killer cell response, while contributing to extravasation of tumor cells, metastatic potential, and neovascularization within the tumor. Recent studies showed that attenuation of immunothrombosis, modulation of platelets and their factors have a good perspective in immunotherapy optimization. Particularly, blockade of intra-tumoral platelet-associated programmed death-ligand 1 might promote anti-tumor T cell-induced cytotoxicity. Collectively, these findings suggest that platelets might represent the source of relevant cancer staging biomarkers, as well as promising targets and carriers in immunotherapeutic approaches for combating cancer.
Collapse
Affiliation(s)
- Drenka Trivanović
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr. Subotica 4, PBOX 102, 11129, Belgrade 11000, Serbia.
| | - Slavko Mojsilović
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr. Subotica 4, PBOX 102, 11129, Belgrade 11000, Serbia
| | | | - Vladimir Jurišić
- Faculty of Medical Sciences, University of Kragujevac, Kragujevac 34000, Serbia
| | - Aleksandra Jauković
- Group for Hematology and Stem Cells, Institute for Medical Research, National Institute of Republic of Serbia, University of Belgrade, Dr. Subotica 4, PBOX 102, 11129, Belgrade 11000, Serbia
| |
Collapse
|
13
|
Li S, Lu Z, Wu S, Chu T, Li B, Qi F, Zhao Y, Nie G. The dynamic role of platelets in cancer progression and their therapeutic implications. Nat Rev Cancer 2024; 24:72-87. [PMID: 38040850 DOI: 10.1038/s41568-023-00639-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/13/2023] [Indexed: 12/03/2023]
Abstract
Systemic antiplatelet treatment represents a promising option to improve the therapeutic outcomes and therapeutic efficacy of chemotherapy and immunotherapy due to the critical contribution of platelets to tumour progression. However, until recently, targeting platelets as a cancer therapeutic has been hampered by the elevated risk of haemorrhagic and thrombocytopenic (low platelet count) complications owing to the lack of specificity for tumour-associated platelets. Recent work has advanced our understanding of the molecular mechanisms responsible for the contribution of platelets to tumour progression and metastasis. This has led to the identification of the biological changes in platelets in the presence of tumours, the complex interactions between platelets and tumour cells during tumour progression, and the effects of platelets on antitumour therapeutic response. In this Review, we present a detailed picture of the dynamic roles of platelets in tumour development and progression as well as their use in diagnosis, prognosis and monitoring response to therapy. We also provide our view on how to overcome challenges faced by the development of precise antiplatelet strategies for safe and efficient clinical cancer therapy.
Collapse
Affiliation(s)
- Suping Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China.
| | - Zefang Lu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Suying Wu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Tianjiao Chu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- College of Pharmaceutical Science, Jilin University, Changchun, China
| | - Bozhao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Feilong Qi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
- Department of Chemistry, Tsinghua University, Beijing, China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|