1
|
Zhang L, Liu J, Qin X, Liu W. Platelet-Acute Leukemia Interactions. Clin Chim Acta 2022; 536:29-38. [PMID: 36122665 DOI: 10.1016/j.cca.2022.09.015] [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: 06/16/2022] [Revised: 09/12/2022] [Accepted: 09/12/2022] [Indexed: 12/01/2022]
Abstract
Acute leukemia (AL) is a hematological malignancy with high morbidity and mortality that is caused by abnormal hematopoietic stem cells. AL can change the parameters, quality, and function of platelets through numerous mechanisms, resulting in bleeding and even death in AL patients. Hence, AL patients are often clinically treated using normal platelet transfusion. However, studies have found that platelets can also affect AL cells. This review discusses the changes occurring in platelet count, mean platelet volume, platelet distribution width, reticulated platelets, platelet membrane glycoprotein, platelet aggregation, and activation in AL patients, the causes of these changes, and the possible significance of these changes for patient prognosis. The effects of platelets on the proliferation and drug resistance of AL cells are also discussed.
Collapse
Affiliation(s)
- Li Zhang
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan 646000, China
| | - Jing Liu
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan 646000, China
| | - Xiang Qin
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan 646000, China
| | - Wenjun Liu
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan 646000, China.
| |
Collapse
|
2
|
Validation of a Mathematical Model Describing the Dynamics of Chemotherapy for Chronic Lymphocytic Leukemia In Vivo. Cells 2022; 11:cells11152325. [PMID: 35954169 PMCID: PMC9367352 DOI: 10.3390/cells11152325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
In recent years, mathematical models have developed into an important tool for cancer research, combining quantitative analysis and natural processes. We have focused on Chronic Lymphocytic Leukemia (CLL), since it is one of the most common adult leukemias, which remains incurable. As the first step toward the mathematical prediction of in vivo drug efficacy, we first found that logistic growth best described the proliferation of fluorescently labeled murine A20 leukemic cells injected in immunocompetent Balb/c mice. Then, we tested the cytotoxic efficacy of Ibrutinib (Ibr) and Cytarabine (Cyt) in A20-bearing mice. The results afforded calculation of the killing rate of the A20 cells as a function of therapy. The experimental data were compared with the simulation model to validate the latter’s applicability. On the basis of these results, we developed a new ordinary differential equations (ODEs) model and provided its sensitivity and stability analysis. There was excellent accordance between numerical simulations of the model and results from in vivo experiments. We found that simulations of our model could predict that the combination of Cyt and Ibr would lead to approximately 95% killing of A20 cells. In its current format, the model can be used as a tool for mathematical prediction of in vivo drug efficacy, and could form the basis of software for prediction of personalized chemotherapy.
Collapse
|
3
|
Investigation of Biomarkers Associated with Low Platelet Counts in Normal Karyotype Acute Myeloid Leukemia. Int J Mol Sci 2022; 23:ijms23147772. [PMID: 35887121 PMCID: PMC9320053 DOI: 10.3390/ijms23147772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 02/05/2023] Open
Abstract
Acute myeloid leukemia (AML) patients are at risk of bleeding due to disease-related lack of platelets and systemic coagulopathy. Platelets play a role in hemostasis. Leukemic blasts have been shown to alter platelet activation in vitro. Here we investigated biomarkers associated with thrombocytopenia in normal karyotype AML (NK-AML). From The Cancer Genome Atlas database, case-control study was performed between normal karyotype (NK) platelet-decreased AML (PD-AML, platelet count < 100 × 109/L, n = 24) and NK platelet-not-decreased AML (PND-AML, with platelet count ≥ 100 × 109/L, n = 13). Differentially expressed gene analysis, pathway analysis and modelling for predicting platelet decrease in AML were performed. DEG analysis and pathway analysis revealed 157 genes and eight pathways specific for PD-AML, respectively. Most of the eight pathways were significantly involved in G-protein-coupled receptor-related pathway, cytokine-related pathway, and bone remodeling pathway. Among the key genes involved in at least one pathway, three genes including CSF1R, TNFSF15 and CLEC10A were selected as promising biomarkers for predicting PD-AML (0.847 of AUC in support vector machine model). This is the first study that identified biomarkers using RNA expression data analysis and could help understand the pathophysiology in AML with low platelet count.
Collapse
|
4
|
Bumbea H, Vladareanu AM, Dumitru I, Popov VM, Ciufu C, Nicolescu A, Onisai M, Marinescu C, Cisleanu D, Voican I, Sarghi S. Platelet Defects in Acute Myeloid Leukemia-Potential for Hemorrhagic Events. J Clin Med 2021; 11:jcm11010118. [PMID: 35011859 PMCID: PMC8745388 DOI: 10.3390/jcm11010118] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/04/2021] [Accepted: 12/21/2021] [Indexed: 11/19/2022] Open
Abstract
Background and objectives: In acute myeloid leukemia (AML), extensive bleeding is one of the most frequent causes of death. Impaired activation and aggregation processes were identified in previous studies on platelet behaviour associated with this disease. This study’s aim was to examine platelet function in correlation with other haemorrhage risk factors (fever, sepsis, recent bleeding, uraemia, leucocytosis, haematocrit value, treatment). Design and methods: The analysis of platelet surface proteins (Glycoprotein Ib-IX (CD42b, CD42a), Glycoprotein IIb-IIIa (CD41, CD61), p-selectin (CD62P), granulophysin (CD63)) was conducted by flowcytometry from samples of whole blood in patients with acute myeloid leukaemia in different stages of diagnosis and therapy (n = 22) in comparison with healthy human controls (n = 10). Results and interpretations: Our results show a significant decrease in fluorescence level associated with platelet activation markers (CD63 (14.11% vs. 40.78 % p < 0.05); CD62P (15.26% vs. 28.23% p < 0.05)); adhesion markers (CD42b (69.08% vs. 84.41% p < 0.05)) and aggregation markers (CD61 (83.79% vs. 98.62% p < 0.001)) in patients compared to controls. The levels of CD41 (80.62% vs. 86.31%, p = 0.290) and CD42a (77.98% vs. 94.15%, p = 0.99) demonstrate no significant differences in the two groups. Conclusion: The AML patients present changes in adhesion receptors and activation markers, suggesting a functional defect or denatured intracellular signalling in platelets. The exposed data indicate that flow cytometry can effectively identify multiple functional platelet impairments in AML pathogenesis.
Collapse
Affiliation(s)
- Horia Bumbea
- Department of Hematology, Emergency University Hospital, 050098 Bucharest, Romania; (H.B.); (A.M.V.); (I.D.); (C.C.); (A.N.); (M.O.); (C.M.); (D.C.); (I.V.)
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Ana Maria Vladareanu
- Department of Hematology, Emergency University Hospital, 050098 Bucharest, Romania; (H.B.); (A.M.V.); (I.D.); (C.C.); (A.N.); (M.O.); (C.M.); (D.C.); (I.V.)
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Ion Dumitru
- Department of Hematology, Emergency University Hospital, 050098 Bucharest, Romania; (H.B.); (A.M.V.); (I.D.); (C.C.); (A.N.); (M.O.); (C.M.); (D.C.); (I.V.)
| | - Viola Maria Popov
- Department of Hematology, Colentina Clinical Hospital, 020125 Bucharest, Romania
- Correspondence:
| | - Cristina Ciufu
- Department of Hematology, Emergency University Hospital, 050098 Bucharest, Romania; (H.B.); (A.M.V.); (I.D.); (C.C.); (A.N.); (M.O.); (C.M.); (D.C.); (I.V.)
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Anca Nicolescu
- Department of Hematology, Emergency University Hospital, 050098 Bucharest, Romania; (H.B.); (A.M.V.); (I.D.); (C.C.); (A.N.); (M.O.); (C.M.); (D.C.); (I.V.)
| | - Minodora Onisai
- Department of Hematology, Emergency University Hospital, 050098 Bucharest, Romania; (H.B.); (A.M.V.); (I.D.); (C.C.); (A.N.); (M.O.); (C.M.); (D.C.); (I.V.)
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Cristina Marinescu
- Department of Hematology, Emergency University Hospital, 050098 Bucharest, Romania; (H.B.); (A.M.V.); (I.D.); (C.C.); (A.N.); (M.O.); (C.M.); (D.C.); (I.V.)
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Diana Cisleanu
- Department of Hematology, Emergency University Hospital, 050098 Bucharest, Romania; (H.B.); (A.M.V.); (I.D.); (C.C.); (A.N.); (M.O.); (C.M.); (D.C.); (I.V.)
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Irina Voican
- Department of Hematology, Emergency University Hospital, 050098 Bucharest, Romania; (H.B.); (A.M.V.); (I.D.); (C.C.); (A.N.); (M.O.); (C.M.); (D.C.); (I.V.)
| | - Sinziana Sarghi
- (VP) Centre, Hospitalier René Dubos, 6 Avenue de l’île de France, 95300 Pontoise, France;
| |
Collapse
|
5
|
Schmied L, Höglund P, Meinke S. Platelet-Mediated Protection of Cancer Cells From Immune Surveillance - Possible Implications for Cancer Immunotherapy. Front Immunol 2021; 12:640578. [PMID: 33777033 PMCID: PMC7988080 DOI: 10.3389/fimmu.2021.640578] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 02/09/2021] [Indexed: 12/19/2022] Open
Abstract
The growing insights in the complex interactions between metastatic cancer-cells and platelets have revealed that platelet tumor cell interactions in the blood stream are an important factor supporting tumor metastasis. An increased coagulability of platelets facilitates the vascular evasion and establishment of solid tumor metastasis. Furthermore, platelets can support an immunosuppressive tumor microenvironment or shield tumor cells directly from engagement of cytotoxic lymphocytes as e.g., natural killer (NK) cells. Platelets are both in the tumor microenvironment and systemically the quantitatively most important source of TGF-β, which is a key cytokine for immunosuppression in the tumor microenvironment. If similar platelet-tumor interactions are of physiological relevance in hematological malignancies remains less well-studied. This might be important, as T- and NK cell mediated graft vs. leukemia effects (GvL) are well-documented and malignant hematological cells have a high exposure to platelets compared to solid tumors. As NK cell-based immunotherapies gain increasing attention as a therapeutic option for patients suffering from hematological and other malignancies, we review the known interactions between platelets and NK cells in the solid tumor setting and discuss how these could also apply to hematological cancers. We furthermore explore the possible implications for NK cell therapy in patients with solid tumors and patients who depend on frequent platelet transfusions. As platelets have a protective and supportive effect on cancer cells, the impact of platelet transfusion on immunotherapy and the combination of immunotherapy with platelet inhibitors needs to be evaluated.
Collapse
Affiliation(s)
- Laurent Schmied
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Petter Höglund
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Stephan Meinke
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
6
|
Ye H, Minhajuddin M, Krug A, Pei S, Chou CH, Culp-Hill R, Ponder J, De Bloois E, Schniedewind B, Amaya ML, Inguva A, Stevens BM, Pollyea DA, Christians U, Grimes HL, D'Alessandro A, Jordan CT. The Hepatic Microenvironment Uniquely Protects Leukemia Cells through Induction of Growth and Survival Pathways Mediated by LIPG. Cancer Discov 2020; 11:500-519. [PMID: 33028621 DOI: 10.1158/2159-8290.cd-20-0318] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 08/11/2020] [Accepted: 10/02/2020] [Indexed: 12/19/2022]
Abstract
Due to the disseminated nature of leukemia, malignant cells are exposed to many different tissue microenvironments, including a variety of extramedullary sites. In the present study, we demonstrate that leukemic cells residing in the liver display unique biological properties and also contribute to systemic changes that influence physiologic responses to chemotherapy. Specifically, the liver microenvironment induces metabolic adaptations via upregulating expression of endothelial lipase in leukemia cells, which not only stimulates tumor cell proliferation through polyunsaturated fatty acid-mediated pathways, but also promotes survival by stabilizing antiapoptotic proteins. Additionally, hepatic infiltration and tissue damage caused by malignant cells induces release of liver-derived enzymes capable of degrading chemotherapy drugs, an event that further protects leukemia cells from conventional therapies. Together, these studies demonstrate a unique role for liver in modulating the pathogenesis of leukemic disease and suggest that the hepatic microenvironment may protect leukemia cells from chemotherapeutic challenge. SIGNIFICANCE: The studies presented herein demonstrate that the liver provides a microenvironment in which leukemia cells acquire unique metabolic properties. The adaptations that occur in the liver confer increased resistance to chemotherapy. Therefore, we propose that therapies designed to overcome liver-specific metabolic changes will yield improved outcomes for patients with leukemia.This article is highlighted in the In This Issue feature, p. 211.
Collapse
Affiliation(s)
- Haobin Ye
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Mohammad Minhajuddin
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anna Krug
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Shanshan Pei
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Chih-Hsing Chou
- Division of Immunobiology, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Rachel Culp-Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jessica Ponder
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Erik De Bloois
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Björn Schniedewind
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Maria L Amaya
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anagha Inguva
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Brett M Stevens
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Daniel A Pollyea
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Uwe Christians
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - H Leighton Grimes
- Division of Immunobiology, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Angelo D'Alessandro
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.,Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Craig T Jordan
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
7
|
Baaten CCFMJ, Moenen FCJI, Henskens YMC, Swieringa F, Wetzels RJH, van Oerle R, Heijnen HFG, Ten Cate H, Holloway GP, Beckers EAM, Heemskerk JWM, van der Meijden PEJ. Impaired mitochondrial activity explains platelet dysfunction in thrombocytopenic cancer patients undergoing chemotherapy. Haematologica 2018; 103:1557-1567. [PMID: 29880611 PMCID: PMC6119160 DOI: 10.3324/haematol.2017.185165] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 06/05/2018] [Indexed: 01/07/2023] Open
Abstract
Severe thrombocytopenia (≤50×109 platelets/L) due to hematological malignancy and intensive chemotherapy is associated with an increased risk of clinically significant bleeding. Since the bleeding risk is not linked to the platelet count only, other hemostatic factors must be involved. We studied platelet function in 77 patients with acute leukemia, multiple myeloma or malignant lymphoma, who experienced chemotherapy-induced thrombocytopenia. Platelets from all patients - independent of disease or treatment type - were to a variable extent compromised in Ca2+ flux, integrin a β activation and P-selectin expression when stimulated with a panelIIbof3 agonists. The patients' platelets were also impaired in spreading on fibrinogen. Whereas the Ca2+ store content was unaffected, the patients' platelets showed ongoing phosphatidylserine exposure, which was not due to apoptotic caspase activity. Interestingly, mitochondrial function was markedly reduced in platelets from a representative subset of patients, as evidenced by a low mitochondrial membrane potential (P<0.001) and low oxygen consumption (P<0.05), while the mitochondrial content was normal. Moreover, the mitochondrial impairments coincided with elevated levels of reactive oxygen species (Spearman's rho=-0.459, P=0.012). Markedly, the impairment of platelet function only appeared after two days of chemotherapy, suggesting origination in the megakaryocytes. In patients with bone marrow recovery, platelet function improved. In conclusion, our findings disclose defective receptor signaling related to impaired mitochondrial bioenergetics, independent of apoptosis, in platelets from cancer patients treated with chemotherapy, explaining the low hemostatic potential of these patients.
Collapse
Affiliation(s)
- Constance C F M J Baaten
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, the Netherlands
| | - Floor C J I Moenen
- Department of Hematology, Maastricht University Medical Centre, the Netherlands
| | - Yvonne M C Henskens
- Central Diagnostic Laboratory, Maastricht University Medical Centre, the Netherlands
| | - Frauke Swieringa
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, the Netherlands.,Department of Protein Dynamics, Leibniz Institute for Analytical Sciences - ISAS-e.V., Dortmund, Germany
| | - Rick J H Wetzels
- Central Diagnostic Laboratory, Maastricht University Medical Centre, the Netherlands
| | - René van Oerle
- Central Diagnostic Laboratory, Maastricht University Medical Centre, the Netherlands.,Laboratory for Clinical Thrombosis and Hemostasis, Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, the Netherlands
| | - Harry F G Heijnen
- Department of Cell Biology and Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, the Netherlands
| | - Hugo Ten Cate
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, the Netherlands.,Laboratory for Clinical Thrombosis and Hemostasis, Department of Internal Medicine, Cardiovascular Research Institute Maastricht, Maastricht University Medical Center, the Netherlands
| | - Graham P Holloway
- Department of Human Health and Nutritional Sciences, University of Guelph, Ontario, Canada
| | - Erik A M Beckers
- Department of Hematology, Maastricht University Medical Centre, the Netherlands
| | - Johan W M Heemskerk
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, the Netherlands
| | - Paola E J van der Meijden
- Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University Medical Centre, the Netherlands
| |
Collapse
|
8
|
Vicente-Ayuso C, Papadopoulos P, Villa-Fajardo M, Prieto B, Álvarez-Carmona AM, Mateo-Morales M, Pérez-López C, Peña-Cortijo A, Polo-Zarzuela M, Sáez I, Martín MP, Benavente-Cuesta C, González-Fernández A, Martínez-Martínez R, Zwaginga JJ, Seghatchian J, Gutiérrez L. Identification of underlying and transfusion-related platelet qualitative alterations in the hemato-oncologic patient. Transfus Apher Sci 2017; 56:756-768. [PMID: 28970003 DOI: 10.1016/j.transci.2017.08.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hemato-oncologic patients with chemotherapy-induced thrombocytopenia are one of the populations receiving platelet transfusions. The general practice with these patients is to give prophylactic platelet transfusions when platelet counts fall below 10×109PLT/L. However, in more than 40% of these patients, platelet transfusion does not prevent bleeding. The reason of the low efficacy of platelet transfusion in the context of chemotherapy patients is not entirely understood. We therefore aimed at immunophenotyping the expression of platelet surface and activation markers and thrombopoietin levels from hemato-oncologic patients before and after transfusion. A more detailed follow-up was performed in three patients that underwent autologous bone marrow transplantation. As previously reported, basal platelet activation was observed in hemato-oncologic patients. Based on flow cytometry parameters, i.e. the percentage of positivity and mean fluorescence intensity (MFI) distribution, our data provide an additional interpretation of platelet acquired qualitative changes in the hemato-oncologic patient. From our results we propose: first, the underlying activation of platelets in the hemato-oncologic patient is accompanied by loss of expression of the platelet receptors that are susceptible to protease-mediated shedding; second, soon after transfusion, the newly circulating donor platelets show additional activation, which may result in subsequent platelet receptor recycling and potential accelerated clearance of these activated platelets. In conclusion, the immunophenotype of circulating platelets changes after prophylactic platelet transfusion. Next to platelet count increment, exploration of this immunophenotype might help to explain transfusion refractory bleeding in hemato-oncologic patients. Eventually this may lead to personalization and improvement of the present platelet transfusion support regime.
Collapse
Affiliation(s)
- Carmen Vicente-Ayuso
- Department of Hematology, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Petros Papadopoulos
- Department of Hematology, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - María Villa-Fajardo
- Department of Hematology, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Blanca Prieto
- Department of Hematology, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | | | - Marta Mateo-Morales
- Servicio de Hematología y Hemoterapia, Hospital Clínico San Carlos, Madrid, Spain
| | - Cristina Pérez-López
- Servicio de Hematología y Hemoterapia, Hospital Clínico San Carlos, Madrid, Spain
| | | | - Marta Polo-Zarzuela
- Servicio de Hematología y Hemoterapia, Hospital Clínico San Carlos, Madrid, Spain
| | - Isabel Sáez
- Servicio de Hematología y Hemoterapia, Hospital Clínico San Carlos, Madrid, Spain
| | - María Paz Martín
- Department of Hematology, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Celina Benavente-Cuesta
- Department of Hematology, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Ataúlfo González-Fernández
- Department of Hematology, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | | | - Jaap Jan Zwaginga
- Center for Clinical Transfusion Research, Sanquin Research, Leiden, The Netherlands; Department of Immunohaematology and Blood Transfusion, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Jerard Seghatchian
- International Consultancy in Blood Components Quality/Safety Improvement and DDR Strategy, London, United Kingdom.
| | - Laura Gutiérrez
- Department of Hematology, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain.
| |
Collapse
|
9
|
Yan M, Jurasz P. The role of platelets in the tumor microenvironment: From solid tumors to leukemia. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:392-400. [PMID: 26193075 DOI: 10.1016/j.bbamcr.2015.07.008] [Citation(s) in RCA: 115] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/06/2015] [Accepted: 07/08/2015] [Indexed: 12/25/2022]
Abstract
Platelets are increasingly being recognized for promoting tumor growth and metastasis. Many cells derived from solid tumors have the ability to aggregate platelets, and this ability correlates with their metastatic potential. Over the past half century, our understanding of tumor cell-induced platelet aggregation (TCIPA) has grown beyond the simple concept that tumor cell-containing microthrombi mechanically embolize the microvasculature. Tumor cell-activated platelets secrete a multitude of factors that reciprocally act on tumor cells, as well as other cells within the tumor microenvironment; thus, affecting both parenychma and tumor-associated stroma. In this review, we summarize the current knowledge of tumor cell-platelet interactions and their influence on the tumor microenvironment, including how these interactions impact neoplastic epithelial cells, endothelial cells, pericytes, fibroblasts, immune cells, and early metastatic niches. In addition, we review the current knowledge of platelet-cancer cell interactions within hematological malignancies and speculate on how platelets may influence the leukemic microenvironment. This article is part of a Special Issue entitled: Tumor Microenvironment Regulation of Cancer Cell Survival, Metastasis, Inflammation, and Immune Surveillance edited by Peter Ruvolo and Gregg L. Semenza.
Collapse
Affiliation(s)
- MengJie Yan
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Paul Jurasz
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Centre, University of Alberta, Edmonton, Alberta, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
10
|
Platelets promote mitochondrial uncoupling and resistance to apoptosis in leukemia cells: a novel paradigm for the bone marrow microenvironment. CANCER MICROENVIRONMENT 2014; 7:79-90. [PMID: 25112275 DOI: 10.1007/s12307-014-0149-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 07/22/2014] [Indexed: 12/17/2022]
Abstract
Here we report that leukemia cell lines and primary CD34+ leukemic blasts exposed to platelet rich plasma (PRP) or platelet lysates (PL) display increased resistance to apoptosis induced by mitochondria-targeted agents ABT-737 and CDDO-Me. Intriguingly, leukemia cells exposed to platelet components demonstrate a reduction in mitochondrial membrane potential (ΔΨM) and a transient increase in oxygen consumption, suggestive of mitochondrial uncoupling. Accompanying the ranolazine-sensitive increase in oxygen consumption, a reduction in triglyceride content was also observed in leukemia cells cultured with platelet components indicating that lipolysis and fatty acid oxidation may support the molecular reduction of oxygen in these cells. Mechanistically, platelet components antagonized Bax oligomerization in accordance with previous observations supporting an antiapoptotic role for fatty acid oxidation in leukemia cells. Lastly, substantiating the notion that mitochondrial uncoupling reduces oxidative stress, platelet components induced a marked decrease in basal and rotenone-induced superoxide levels in leukemia cells. Taken together, the decrease in ΔΨM, the transient increase in ranolazine-sensitive oxygen consumption, the reduction in triglyceride levels, and the reduced generation of superoxide, all accompanying the increased resistance to mitochondrial apoptosis, substantiate the hypothesis that platelets may contribute to the chemoprotective sanctuary of the bone marrow microenvironment via promotion of mitochondrial uncoupling.
Collapse
|
11
|
Abstract
Acute leukemia is a hematopoietic stem cell malignant disease, with abnormal proliferation of leukemic and immature cells that suppress the production of normal blood cells and extensively invade peripheral tissues. The bleeding complications are very common in acute leukemia and often lead to death. One major cause for hemorrhage is thrombocytopenia, which is caused by the replacement of normal bone marrow cells with leukemic cells and the inhibition of megakaryocytes functions. Declines in platelet count as well as in function in acute leukemia have been reported in many studies. Here, we reviewed the literatures concerning platelet changes in acute leukemia.
Collapse
Affiliation(s)
- Xu Qian
- Department of Pediatrics, Affiliated Hospital of Luzhou Medical College, Luzhou, 646000, Sichuan, China,
| | | |
Collapse
|
12
|
Bruserud Ø. Bidirectional crosstalk between platelets and monocytes initiated by Toll-like receptor: an important step in the early defense against fungal infections? Platelets 2012; 24:85-97. [PMID: 22646762 DOI: 10.3109/09537104.2012.678426] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Monocytes are important in the defense against fungal infections due to their phagocytic and immunoregulatory functions. Platelets also contribute in such immune responses through their release of soluble mediators, including chemokines as well as several other soluble mediators. Both monocytes and platelets express several Toll-like receptors (TLRs) that can recognize fungal molecules and thus initiate intracellular signaling events. TLR ligation on monocytes and platelets may thereby be an early immunological event and function as an initiator of a local proinflammatory crosstalk between platelets and monocytes resulting in (i) monocyte-induced increase of platelet activation and (ii) platelet-associated enhancement of the monocyte activation/function. These effects may have clinical implications both for the efficiency of antifungal treatment and for the predisposition to fungal infections, for example, increased predisposition in patients with thrombocytopenia/monocytopenia due to chemotherapy- or disease-induced bone marrow failure.
Collapse
Affiliation(s)
- Øyvind Bruserud
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
13
|
Cytosine arabinoside-induced cytogenotoxicity in bone marrow and spermatogonial cells of mice and its potential transmission through the male germline. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2009; 673:29-36. [DOI: 10.1016/j.mrgentox.2008.11.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2008] [Revised: 11/11/2008] [Accepted: 11/16/2008] [Indexed: 11/20/2022]
|
14
|
Mills PJ, Parker B, Jones V, Adler KA, Perez CJ, Johnson S, Cohen-Zion M, Marler M, Sadler GR, Dimsdale JE, Ancoli-Israel S. The effects of standard anthracycline-based chemotherapy on soluble ICAM-1 and vascular endothelial growth factor levels in breast cancer. Clin Cancer Res 2005; 10:4998-5003. [PMID: 15297400 DOI: 10.1158/1078-0432.ccr-0734-04] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The circulating soluble form of intercellular adhesion molecule-1 (sICAM-1) and vascular endothelial growth factor (VEGF) are elevated in women with breast cancer and associated with tumor progression and poor prognosis. This study examined the effects of anthracycline-based chemotherapy on plasma sICAM-1 and VEGF, as well as soluble P-selectin, von Willebrand factor, and interleukin-6 levels. EXPERIMENTAL DESIGN Twenty-six women diagnosed with stage I-IIIA breast cancer (mean age, 48.4 +/- 10.4 years; range, 34-79 years) were studied before (week 1) and at weeks 2 and 3 of cycles 1 and 4 of chemotherapy. RESULTS The initial effect of chemotherapy was to reduce sICAM-1 levels; compared with pretreatment, sICAM-1 levels were decreased at week 2 of both cycles (P values < 0.01). sICAM-1 levels were elevated, however, at the start of cycle 4 as compared with pretreatment (P < 0.01). Chemotherapy led to an increase in sICAM-1 levels in node-positive but not node-negative patients (P < 0.01). VEGF levels were decreased at week 2 of cycle 4 (P = 0.001) and remained so at week 3. Similar to sICAM-1, VEGF levels were elevated at the start of cycle 4 as compared with pretreatment (P < 0.006). Soluble P-selectin levels decreased during week 2 of cycle 4 (P = 0.026). Neither interleukin-6 or von Willebrand factor were significantly changed in response to chemotherapy. CONCLUSIONS The findings support prior studies suggesting that sICAM-1 levels derive from sources other than endothelial cells. In addition, whereas the more immediate effect of chemotherapy is to reduce sICAM-1 and VEGF, continued treatment may lead to significant elevations.
Collapse
Affiliation(s)
- Paul J Mills
- Department of Psychiatry, University of California San Diego, San Diego, California 92103, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Leinoe EB, Hoffmann MH, Kjaersgaard E, Johnsen HE. Multiple platelet defects identified by flow cytometry at diagnosis in acute myeloid leukaemia. Br J Haematol 2004; 127:76-84. [PMID: 15384980 DOI: 10.1111/j.1365-2141.2004.05156.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Summary Previous findings of megakaryocytic hypogranulation and dysmegakaryocytopoietic features in acute myeloid leukaemia (AML) strongly indicate defects in platelet production. The bleeding tendency of these patients may result from dysregulated platelet production, resulting in thrombocytopenia as well as qualitative platelet defects. The present study examined platelet function at diagnosis in 50 AML patients by whole blood flow cytometry. Following in vitro platelet agonist stimulation, platelet activation markers were analysed and compared with 20 healthy individuals. To detect recent in vivo platelet activation, plasma soluble P-selectin (sP-selectin) was measured. Flow cytometric analysis of platelet activation markers demonstrated reduced CD62P [35.6 vs. 118.5 x 10(3) molecules of equivalent soluble fluorochrome (MESF); P < 0.0001], CD63 (11.3 vs. 50.7 x 10(3) MESF; P < 0.0001), and PAC-1 (41.5 vs. 90.5%; P = 0.0001) while reductions in CD42b were abnormal (45.6 vs. 70%; P < 0.0001). sP-selectin levels were similar in patients and healthy controls (0.04 vs. 0.27 fg/platelet; P = 0.84). The presented data indicate that AML pathogenesis may result in multiple platelet defects, involving adhesion, aggregation, and secretion and demonstrate that flow cytometry is a feasible method for platelet function analysis in patients with thrombocytopenia.
Collapse
Affiliation(s)
- E B Leinoe
- The Research Laboratory, Department of Haematology, Herlev University Hospital, Copenhagen, Denmark.
| | | | | | | |
Collapse
|
16
|
Sakurai Y, Shimojima M, Miyazawa T, Masuoka K, Tohya Y, Akashi H. Identification of the feline CD63 homologue using retrovirus-mediated expression cloning. Vet Immunol Immunopathol 2004; 98:185-91. [PMID: 15010227 DOI: 10.1016/j.vetimm.2003.12.005] [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: 06/12/2003] [Revised: 10/31/2003] [Accepted: 12/02/2003] [Indexed: 10/26/2022]
Abstract
Recently, we combined a retrovirus-mediated expression cloning with a simple screening method using non-adherent cells and panning [Anal. Biochem. 315 (2003) 138]. In this study, we applied this method to identify the antigen recognized by an uncharacterized monoclonal antibody raised against a feline cell line, and identified it as the feline homologue of CD63. This simple method is useful for characterizing unknown antibodies that recognize cell surface molecules. Furthermore, the monoclonal antibody identified as an anti-feline CD63 antibody will be useful for studying feline molecular function(s).
Collapse
Affiliation(s)
- Yumiko Sakurai
- Department of Veterinary Microbiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | | | | | | | | | | |
Collapse
|
17
|
Glenjen N, Mosevoll KA, Bruserud Ø. Serum levels of angiogenin, basic fibroblast growth factor and endostatin in patients receiving intensive chemotherapy for acute myelogenous leukemia. Int J Cancer 2002; 101:86-94. [PMID: 12209593 DOI: 10.1002/ijc.10566] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Angiogenesis seems to be important both in the pathogenesis of acute myelogenous leukemia (AML) and for the susceptibility of AML blasts to chemotherapy. Recent clinical studies even suggest that antiangiogenic therapy can induce disease control in patients with AML relapse. In this context we have investigated the profile of the systemic component of angiogenic regulation in AML by characterizing the serum levels of (i) the angiogenic regulators angiogenin, basic fibroblast growth factor (bFGF) and endostatin; (ii) the endothelial cell marker soluble (s) E-selectin. Patients with untreated AML had increased levels of angiogenin, endostatin and sE-selectin, whereas the levels of bFGF were not significantly altered. The systemic levels of the proangiogenic bFGF, the antiangiogenic endostatin and the endothelial cell marker sE-selectin showed significant correlations, whereas angiogenin and sE-selectin levels were not correlated. Furthermore, intensive chemotherapy resulted in decreased systemic levels of the 2 proangiogenic mediators angiogenin and bFGF, whereas endostatin levels remained high after treatment. Although angiogenin normally is a part of the acute phase reaction, its systemic levels were not altered when patients with chemotherapy-induced cytopenia developed complicating bacterial infections. Our results suggest that intensive chemotherapy can modulate the systemic component of angiogenic regulation in AML patients.
Collapse
Affiliation(s)
- Nils Glenjen
- Section for Hematology, Department of Medicine, Haukeland University Hospital and the University of Bergen, Norway.
| | | | | |
Collapse
|