1
|
Choi JH, Kim K. Polyhexamethylene Guanidine Phosphate Enhanced Procoagulant Activity through Oxidative-Stress-Mediated Phosphatidylserine Exposure in Platelets. TOXICS 2024; 12:50. [PMID: 38251006 PMCID: PMC10820372 DOI: 10.3390/toxics12010050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/23/2024]
Abstract
Polyhexamethylene guanidine phosphate (PHMG-p) is a common biocidal disinfectant that is widely used in industry and household products. However, PHMG-p was misused as a humidifier disinfectant (HD) in South Korea, which had fatal health effects. Various health problems including cardiovascular diseases were observed in HD-exposed groups. However, the potential underlying mechanism of HD-associated cardiovascular diseases is poorly understood. Here, we examined the procoagulant activity of platelets caused by PHMG-p and clarified the underlying mechanism. PHMG-p enhanced phosphatidylserine (PS) exposure through alteration of phospholipid transporters, scramblase, and flippase. Intracellular calcium elevation, intracellular ATP depletion, and caspase-3 activation appeared to underlie phospholipid transporter dysregulation caused by PHMG-p, which was mediated by oxidative stress and mitochondrial dysfunction. Notably, antioxidant enzyme catalase and calcium chelator EGTA reversed PHMG-p-induced PS exposure and thrombin generation, confirming the contributive role of oxidative stress and intracellular calcium in the procoagulant effects of PHMG-p. These series of events led to procoagulant activation of platelets, which was revealed as enhanced thrombin generation. Collectively, PHMG-p triggered procoagulant activation of platelets, which may promote prothrombotic risks and cardiovascular diseases. These findings improve our understanding of HD-associated cardiovascular diseases.
Collapse
Affiliation(s)
| | - Keunyoung Kim
- College of Pharmacy, Kangwon National University, Chuncheon 24341, Republic of Korea;
| |
Collapse
|
2
|
Liang G, Li X, Xu Q, Yang Z, Li J, Yang T, Wang G, Lei H. Development and validation of a nomogram model for predicting the risk of venous thromboembolism in lymphoma patients undergoing chemotherapy: a prospective cohort study conducted in China. Ann Med 2023; 55:2275665. [PMID: 38132496 PMCID: PMC10763890 DOI: 10.1080/07853890.2023.2275665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/19/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND The mechanism of Venous thromboembolism (VTE) is complicated and difficult to prevent due to factors such as bone marrow invasion, therapy, and immune-mediated effects. This study aims to establish a nomogram model for predicting the risk of thrombosis in lymphoma patients undergoing chemotherapy, which has been increasing over the past 30 years. METHODS The data of lymphoma patients from the Affiliated Cancer Hospital of Chongqing University in China between 2018 and 2020 were analyzed. This included age, sex, body mass index, ECOG score, histological type, Ann Arbour Stage, white blood cells count, haemoglobin level, platelet count, D-dimer level, and chemotherapy cycle. Univariate and multivariate cox analysis was used to determine the risk factors for VTE. Characteristic variables were selected to construct a nomogram model which was then evaluated using ROC curve and calibration. RESULTS Age, sex, PLT, D-dimer and chemotherapy cycle were considered as independent influencing factors of VTE. The mean (standard deviation) of the C index, AUC and Royston D statistics of 1000 cross-validations of the Nomogram model were 0.78 (0.01), 0.81 (0.01) and 1.61(0.07), respectively. It indicates a good calibration degree and applicability value as shown by the calibration curve. The DCA curve showed a rough threshold range of 0.05-0.60 with a good model. CONCLUSIONS We have established and validated a nomogram model for predicting the risk of thrombosis in lymphoma patients. This model can assess the risk of thrombosis in each individual patient, enabling the identification of high-risk groups and targeted preventive treatment.
Collapse
Affiliation(s)
- Guanzhong Liang
- Chongqing Cancer Multi-omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaosheng Li
- Chongqing Cancer Multi-omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Qianjie Xu
- Department of Health Statistics, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Zailin Yang
- Chongqing Cancer Multi-omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Jieping Li
- Chongqing Cancer Multi-omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Tao Yang
- Chongqing Cancer Multi-omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Guixue Wang
- MOE Key Lab for Biorheological Science and Technology, State and Local Joint Engineering Laboratory for Vascular Implants, College of Bioengineering Chongqing University, Chongqing, China
| | - Haike Lei
- Chongqing Cancer Multi-omics Big Data Application Engineering Research Center, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
3
|
Fuentes E, Arauna D, Araya-Maturana R. Regulation of mitochondrial function by hydroquinone derivatives as prevention of platelet activation. Thromb Res 2023; 230:55-63. [PMID: 37639783 DOI: 10.1016/j.thromres.2023.08.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/07/2023] [Accepted: 08/18/2023] [Indexed: 08/31/2023]
Abstract
Platelet activation plays an essential role in the pathogenesis of thrombotic events in different diseases (e.g., cancer, type 2 diabetes, Alzheimer's, and cardiovascular diseases, and even in patients diagnosed with coronavirus disease 2019). Therefore, antiplatelet therapy is essential to reduce thrombus formation. However, the utility of current antiplatelet drugs is limited. Therefore, identifying novel antiplatelet compounds is very important in developing new drugs. In this context, the involvement of mitochondrial function as an efficient energy source required for platelet activation is currently accepted; however, its contribution as an antiplatelet target still has little been exploited. Regarding this, the intramolecular hydrogen bonding of hydroquinone derivatives has been described as a structural motif that allows the reach of small molecules at mitochondria, which can exert antiplatelet activity, among others. In this review, we describe the role of mitochondrial function in platelet activation and how hydroquinone derivatives exert antiplatelet activity through mitochondrial regulation.
Collapse
Affiliation(s)
- Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, MIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Universidad de Talca, Talca 3480094, Chile.
| | - Diego Arauna
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, MIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Universidad de Talca, Talca 3480094, Chile
| | - Ramiro Araya-Maturana
- Instituto de Química de Recursos Naturales, MIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Universidad de Talca, Talca 3460000, Chile
| |
Collapse
|
4
|
Zhao J, Shi Y, Xue L, Liang Y, Shen J, Wang J, Wu M, Chen H, Kong M. Glucose-decorated engineering platelets for active and precise tumor-targeted drug delivery. Biomater Sci 2023; 11:3965-3975. [PMID: 37114937 DOI: 10.1039/d3bm00326d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Precise targeted delivery of therapeutic agents is crucial for tumor therapy. As an emerging fashion, cell-based delivery provides better biocompatibility and lower immunogenicity and enables a more precise accumulation of drugs in tumor cells. In this study, a novel engineering platelet was constructed through cell membrane fusion with a synthesized glycolipid molecule, DSPE-PEG-Glucose (DPG). The obtained glucose-decorated platelets (DPG-PLs) maintained their resting state with structural and functional integrities, while they would be activated and triggered to release their payloads once they arrive at the tumor microenvironment. Glucose decoration was verified to impart the DPG-PLs with stronger binding effects toward tumor cells that overexpress GLUT1 on their surfaces. Together with the natural homing property toward tumor sites and bleeding injury, doxorubicin (DOX)-loaded platelets (DPG-PL@DOX) exhibited the strongest antitumor effects on a mouse melanoma model, and the antitumor effect was significantly enhanced in the tumor bleeding model. DPG-PL@DOX provides an active and precise solution for tumor-targeted drug delivery, especially for postoperative treatments.
Collapse
Affiliation(s)
- Jiaxuan Zhao
- College of Marine Life Science, Ocean University of China, 5 Yushan Road, 266003, Qingdao, China.
| | - Yan Shi
- College of Marine Life Science, Ocean University of China, 5 Yushan Road, 266003, Qingdao, China.
| | - Lixia Xue
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 227 Chongqing South Road, 200025, Shanghai, China.
| | - Yuqing Liang
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 227 Chongqing South Road, 200025, Shanghai, China.
| | - Jiale Shen
- College of Marine Life Science, Ocean University of China, 5 Yushan Road, 266003, Qingdao, China.
| | - Jiarui Wang
- College of Marine Life Science, Ocean University of China, 5 Yushan Road, 266003, Qingdao, China.
| | - Meng Wu
- College of Marine Life Science, Ocean University of China, 5 Yushan Road, 266003, Qingdao, China.
| | - Hao Chen
- Department of Neurosurgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 227 Chongqing South Road, 200025, Shanghai, China.
| | - Ming Kong
- College of Marine Life Science, Ocean University of China, 5 Yushan Road, 266003, Qingdao, China.
| |
Collapse
|
5
|
Sirikul W, Buawangpong N, Pruksakorn D, Charoentum C, Teeyakasem P, Koonrungsesomboon N. The Survival Outcomes, Prognostic Factors and Adverse Events following Systemic Chemotherapy Treatment in Bone Sarcomas: A Retrospective Observational Study from the Experience of the Cancer Referral Center in Northern Thailand. Cancers (Basel) 2023; 15:cancers15071979. [PMID: 37046640 PMCID: PMC10092999 DOI: 10.3390/cancers15071979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/15/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023] Open
Abstract
This study aimed to assess survival outcomes, prognostic factors, and adverse events following chemotherapy treatment for osteosarcoma and Ewing’s sarcoma. This retrospective observational study was conducted to collect the data of the patients with osteosarcoma or Ewing’s sarcoma who received chemotherapy treatment between 2008 and 2019. The flexible parametric survival model was performed to explore the adjusted survival probability and the prognostic factors. A total of 102 patients (79 with osteosarcoma and 23 with Ewing’s sarcoma) were included. The estimated 5-year disease-free survival (DFS) and 5-year overall survival (OS) probabilities in patients with resectable disease were 60.9% and 63.3% for osteosarcoma, and 54.4% and 88.3% for Ewing’s sarcoma, respectively, whereas the 5-year DFS and 5-year OS for those with unresectable/metastatic disease remained below 25%. Two prognostic factors for osteosarcoma included a response to neoadjuvant chemotherapy and female gender. Ewing’s sarcoma patients aged 25 years and older were significantly associated with poorer survival outcomes. Of 181 chemotherapy treatment cycles, common self-reported adverse symptoms included tumor pain (n = 32, 17.7%), fever (n = 21, 11.6%), and fatigue (n = 16, 8.8%), while common grade III adverse events included febrile neutropenia (n = 13, 7.3%) and neutropenia (n = 9, 5.1%). There was no chemotherapy-related mortality (grade V) or anaphylaxis events.
Collapse
Affiliation(s)
- Wachiranun Sirikul
- Department of Community Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nida Buawangpong
- Department of Family Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Dumnoensun Pruksakorn
- Department of Orthopedic, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chaiyut Charoentum
- Division of Oncology, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Pimpisa Teeyakasem
- Department of Orthopedic, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nut Koonrungsesomboon
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Clinical Research Center for Food and Herbal Product Trials and Development (CR-FAH), Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Correspondence: ; Tel.: +66-5393-5353
| |
Collapse
|
6
|
Ma W, Rousseau Z, Slavkovic S, Shen C, Yousef GM, Ni H. Doxorubicin-Induced Platelet Activation and Clearance Relieved by Salvianolic Acid Compound: Novel Mechanism and Potential Therapy for Chemotherapy-Associated Thrombosis and Thrombocytopenia. Pharmaceuticals (Basel) 2022; 15:1444. [PMID: 36558895 PMCID: PMC9788583 DOI: 10.3390/ph15121444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/04/2022] [Accepted: 11/15/2022] [Indexed: 11/24/2022] Open
Abstract
Doxorubicin (Dox) is a widely utilized chemotherapeutic; however, it carries side effects, including drug-induced immune thrombocytopenia (DITP) and increased risk of venous thromboembolism (VTE). Currently, the mechanisms for Dox-associated DITP and VTE are poorly understood, and an effective inhibitor to relieve these complications remains to be developed. In this study, we found that Dox significantly induced platelet activation and enhanced platelet phagocytosis by macrophages and accelerated platelet clearance. Importantly, we determined that salvianolic acid C (SAC), a water-soluble compound derived from Danshen root traditionally used to treat cardiovascular diseases, inhibited Dox-induced platelet activation more effectively than current standard-of-care anti-platelet drugs aspirin and ticagrelor. Mechanism studies with tyrosine kinase inhibitors indicate contributions of phospholipase C, spleen tyrosine kinase, and protein kinase C signaling pathways in Dox-induced platelet activation. We further demonstrated that Dox enhanced platelet-cancer cell interaction, which was ameliorated by SAC. Taken together, these findings suggest SAC may be a promising therapy to reduce the risk of Dox-induced DITP, VTE, and the repercussions of amplified platelet-cancer interaction in the tumor microenvironment.
Collapse
Affiliation(s)
- Wenjing Ma
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto Platelet Immunobiology Group, Toronto, ON M5B 1W8, Canada
| | - Zackary Rousseau
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto Platelet Immunobiology Group, Toronto, ON M5B 1W8, Canada
| | - Sladjana Slavkovic
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto Platelet Immunobiology Group, Toronto, ON M5B 1W8, Canada
| | - Chuanbin Shen
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto Platelet Immunobiology Group, Toronto, ON M5B 1W8, Canada
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - George M. Yousef
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto Platelet Immunobiology Group, Toronto, ON M5B 1W8, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Heyu Ni
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Laboratory Medicine, LKSKI-Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto Platelet Immunobiology Group, Toronto, ON M5B 1W8, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Canadian Blood Services Centre for Innovation, Toronto, ON M5G 2M1, Canada
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada
| |
Collapse
|
7
|
Antoniak S, Phungphong S, Cheng Z, Jensen BC. Novel Mechanisms of Anthracycline-Induced Cardiovascular Toxicity: A Focus on Thrombosis, Cardiac Atrophy, and Programmed Cell Death. Front Cardiovasc Med 2022; 8:817977. [PMID: 35111832 PMCID: PMC8801506 DOI: 10.3389/fcvm.2021.817977] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 12/23/2021] [Indexed: 01/13/2023] Open
Abstract
Anthracycline antineoplastic agents such as doxorubicin are widely used and highly effective component of adjuvant chemotherapy for breast cancer and curative regimens for lymphomas, leukemias, and sarcomas. The primary dose-limiting adverse effect of anthracyclines is cardiotoxicity that typically manifests as cardiomyopathy and can progress to the potentially fatal clinical syndrome of heart failure. Decades of pre-clinical research have explicated the complex and multifaceted mechanisms of anthracycline-induced cardiotoxicity. It is well-established that oxidative stress contributes to the pathobiology and recent work has elucidated important central roles for direct mitochondrial injury and iron overload. Here we focus instead on emerging aspects of anthracycline-induced cardiotoxicity that may have received less attention in other recent reviews: thrombosis, myocardial atrophy, and non-apoptotic programmed cell death.
Collapse
Affiliation(s)
- Silvio Antoniak
- Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, United States
- Blood Research Center, University of North Carolina School of Medicine, Chapel Hill, NC, United States
- *Correspondence: Silvio Antoniak
| | - Sukanya Phungphong
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, United States
| | - Zhaokang Cheng
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, United States
- Zhaokang Cheng
| | - Brian C. Jensen
- Cardiology Division, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, NC, United States
- Department of Pharmacology, University of North Carolina School of Medicine, Chapel Hill, NC, United States
- McAllister Heart Institute, University of North Carolina School of Medicine, Chapel Hill, NC, United States
| |
Collapse
|
8
|
Hsu PY, Mammadova A, Benkirane-Jessel N, Désaubry L, Nebigil CG. Updates on Anticancer Therapy-Mediated Vascular Toxicity and New Horizons in Therapeutic Strategies. Front Cardiovasc Med 2021; 8:694711. [PMID: 34386529 PMCID: PMC8353082 DOI: 10.3389/fcvm.2021.694711] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/18/2021] [Indexed: 12/11/2022] Open
Abstract
Vascular toxicity is a frequent adverse effect of current anticancer chemotherapies and often results from endothelial dysfunction. Vascular endothelial growth factor inhibitors (VEGFi), anthracyclines, plant alkaloids, alkylating agents, antimetabolites, and radiation therapy evoke vascular toxicity. These anticancer treatments not only affect tumor vascularization in a beneficial manner, they also damage ECs in the heart. Cardiac ECs have a vital role in cardiovascular functions including hemostasis, inflammatory and coagulation responses, vasculogenesis, and angiogenesis. EC damage can be resulted from capturing angiogenic factors, inhibiting EC proliferation, survival and signal transduction, or altering vascular tone. EC dysfunction accounts for the pathogenesis of myocardial infarction, atherothrombosis, microangiopathies, and hypertension. In this review, we provide a comprehensive overview of the effects of chemotherapeutic agents on vascular toxicity leading to hypertension, microvascular rarefaction thrombosis and atherosclerosis, and affecting drug delivery. We also describe the potential therapeutic approaches such as vascular endothelial growth factor (VEGF)-B and prokineticin receptor-1 agonists to maintain endothelial function during or following treatments with chemotherapeutic agents, without affecting anti-tumor effectiveness.
Collapse
Affiliation(s)
| | | | | | | | - Canan G. Nebigil
- INSERM UMR 1260, Regenerative Nanomedicine, University of Strasbourg, FMTS (Fédération de Médecine Translationnelle de l'Université de Strasbourg), Strasbourg, France
| |
Collapse
|
9
|
Chakraborty R, Rybicki L, Valent J, Garcia AVM, Faiman BM, Khouri J, Samaras CJ, Anwer F, Khorana AA. Arterial thromboembolism in multiple myeloma in the context of modern anti-myeloma therapy. Blood Cancer J 2021; 11:121. [PMID: 34172719 PMCID: PMC8233391 DOI: 10.1038/s41408-021-00513-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/10/2021] [Accepted: 05/21/2021] [Indexed: 02/06/2023] Open
Affiliation(s)
- Rajshekhar Chakraborty
- Multiple Myeloma and Amyloidosis Program, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
| | - Lisa Rybicki
- Blood and Marrow Transplant Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jason Valent
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Alex V Mejia Garcia
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Beth M Faiman
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jack Khouri
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Christy J Samaras
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Faiz Anwer
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Alok A Khorana
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
10
|
Grover SP, Hisada YM, Kasthuri RS, Reeves BN, Mackman N. Cancer Therapy-Associated Thrombosis. Arterioscler Thromb Vasc Biol 2021; 41:1291-1305. [PMID: 33567864 DOI: 10.1161/atvbaha.120.314378] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Steven P Grover
- UNC Blood Research Center, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill
| | - Yohei M Hisada
- UNC Blood Research Center, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill
| | - Raj S Kasthuri
- UNC Blood Research Center, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill
| | - Brandi N Reeves
- UNC Blood Research Center, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill
| | - Nigel Mackman
- UNC Blood Research Center, Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill
| |
Collapse
|
11
|
Hao Z, Lv H, Tan R, Yang X, Liu Y, Xia YL. A Three-Dimensional Microfluidic Device for Monitoring Cancer and Chemotherapy-Associated Platelet Activation. ACS OMEGA 2021; 6:3164-3172. [PMID: 33553932 PMCID: PMC7860090 DOI: 10.1021/acsomega.0c05572] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 01/13/2021] [Indexed: 05/04/2023]
Abstract
Platelet activation and the risk of thrombosis are increased in cancer patients, especially after chemotherapy. Our previous studies indicated that chemotherapy-induced platelet activation is largely due to endothelial cell damage. Thus, simple in vitro tests, such as aggregometry, are not desirable tests to predict platelet responsiveness to different chemotherapeutic agents because other contributory factors, such as tumor cells, endothelial cells, and the flow rate of platelets, also contribute to the formation of cancer-associated thrombosis. Therefore, developing a platelet detection system, which includes all possible risk parameters, is necessary. In the present study, we described a microengineered microfluidic system that contained a drug concentration generator, cancer cell culture chip, and three-dimensional (3D) circular microvascular model covered with a confluent endothelial layer and perfused with human platelets at a stable flow rate. Doxorubicin was injected through two injection sites. Endothelial cell injury was evaluated by counting, cell cytoskeleton observation, and the level of IACM1 and ET-1 in endothelial cells or a culture medium. Prestained platelets were perfused into the artificial blood vessel, and platelet-endothelial cell adhesion was measured. We found that (i) MCF7 cell-released factors had a cytotoxicity effect on both endothelial cells and platelets. (ii) We confirmed that doxorubicin-induced platelet activation was endothelial cell-dependent. (iii) A lower dosage of doxorubicin (0-2.0 μM) induced platelet activation, while a higher dosage of doxorubicin (2.0-4.0 μM) led to platelet death. Our findings indicated that platelet-endothelial cell adhesion could be used as a diagnostic marker of platelet activation, providing a simple and rapid detective way to predict platelet responsiveness before or during chemotherapy.
Collapse
Affiliation(s)
- Zhujing Hao
- Institute
of Cardiovascular Diseases, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
| | - Haichen Lv
- Department
of Cardiology, The First Affiliated Hospital
of Dalian Medical University, Dalian 116000, China
| | - Ruopeng Tan
- Institute
of Cardiovascular Diseases, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
| | - Xiaolei Yang
- Institute
of Cardiovascular Diseases, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
| | - Yang Liu
- Institute
of Cardiovascular Diseases, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
- . Tel: 86-411-83635963-2287
| | - Yun-Long Xia
- Institute
of Cardiovascular Diseases, The First Affiliated
Hospital of Dalian Medical University, Dalian 116000, China
- Department
of Cardiology, The First Affiliated Hospital
of Dalian Medical University, Dalian 116000, China
- . Tel: 86-411-83635963-3004
| |
Collapse
|
12
|
Chen R, Liu X, Law AD, Kanfar S, Maze D, Chan SM, Gupta V, Yee KW, Minden MD, Schimmer AD, Schuh AC, McNamara CJ, Murphy T, Xu A, Falcone U, Seki J, Sibai H. Risk of Thrombosis in Adult Philadelphia-Positive ALL Treated with an Asparaginase-Free ALL Regimen. Curr Oncol 2020; 28:128-137. [PMID: 33704181 PMCID: PMC7816186 DOI: 10.3390/curroncol28010016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 12/15/2020] [Accepted: 12/17/2020] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND venous thromboembolism (VTE) is a well-known complication in adults with acute lymphoblastic leukemia (ALL), especially in patients treated with asparaginase (ASNase)-including regiments. However, VTE risk in adult Philadelphia-positive ALL (Ph+ve ALL) patients treated with non-hyperCVAD chemotherapy is unclear. In this study, we examined VTE incidence in adult Ph+ve ALL patients treated with imatinib plus a pediatric-inspired asparaginase (ASNase)-free regimen modified from the Dana Farber Cancer Institute (DFCI) ALL protocol. METHODS a single centre retrospective review of Ph+ve ALL patients treated at Princess Margaret Cancer Center (PMCC) from 2008-2019 with imatinib plus modified DFCI protocol was conducted. RESULTS of the 123 patients included, 30 (24.3%) had at least 1 radiology confirmed VTE event from diagnosis to the end of maintenance therapy. 86.7% (26/30) of the VTE events occurred during active treatment. Of all VTE events, the majority (53.3%) were DVT and/or PE while another significant portion were catheter-related (40.0%). Major bleeding was observed in 1 patient on VTE treatment with low molecular weight heparin (LMWH). CONCLUSION a high VTE incidence (24.3%) was observed in adults Ph+ve ALL patients treated with imatinib plus an ASNase-free modified DFCI pediatric ALL protocol, suggesting prophylactic anticoagulation should be considered for all adult Ph+ve ALL patients including those treated with ASNase-free regimens.
Collapse
Affiliation(s)
- Ruiqi Chen
- Princess Margaret Cancer Centre, Department of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C1, Canada; (R.C.); (A.D.L.); (S.K.); (D.M.); (S.M.C.); (V.G.); (K.W.Y.); (M.D.M.); (A.D.S.); (A.C.S.); (C.J.M.); (T.M.); (J.S.)
| | - Xing Liu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada; (X.L.); (A.X.)
- Department of Pharmacy, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Arjun D. Law
- Princess Margaret Cancer Centre, Department of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C1, Canada; (R.C.); (A.D.L.); (S.K.); (D.M.); (S.M.C.); (V.G.); (K.W.Y.); (M.D.M.); (A.D.S.); (A.C.S.); (C.J.M.); (T.M.); (J.S.)
| | - Solaf Kanfar
- Princess Margaret Cancer Centre, Department of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C1, Canada; (R.C.); (A.D.L.); (S.K.); (D.M.); (S.M.C.); (V.G.); (K.W.Y.); (M.D.M.); (A.D.S.); (A.C.S.); (C.J.M.); (T.M.); (J.S.)
| | - Dawn Maze
- Princess Margaret Cancer Centre, Department of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C1, Canada; (R.C.); (A.D.L.); (S.K.); (D.M.); (S.M.C.); (V.G.); (K.W.Y.); (M.D.M.); (A.D.S.); (A.C.S.); (C.J.M.); (T.M.); (J.S.)
| | - Steven M. Chan
- Princess Margaret Cancer Centre, Department of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C1, Canada; (R.C.); (A.D.L.); (S.K.); (D.M.); (S.M.C.); (V.G.); (K.W.Y.); (M.D.M.); (A.D.S.); (A.C.S.); (C.J.M.); (T.M.); (J.S.)
| | - Vikas Gupta
- Princess Margaret Cancer Centre, Department of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C1, Canada; (R.C.); (A.D.L.); (S.K.); (D.M.); (S.M.C.); (V.G.); (K.W.Y.); (M.D.M.); (A.D.S.); (A.C.S.); (C.J.M.); (T.M.); (J.S.)
| | - Karen W. Yee
- Princess Margaret Cancer Centre, Department of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C1, Canada; (R.C.); (A.D.L.); (S.K.); (D.M.); (S.M.C.); (V.G.); (K.W.Y.); (M.D.M.); (A.D.S.); (A.C.S.); (C.J.M.); (T.M.); (J.S.)
| | - Mark D. Minden
- Princess Margaret Cancer Centre, Department of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C1, Canada; (R.C.); (A.D.L.); (S.K.); (D.M.); (S.M.C.); (V.G.); (K.W.Y.); (M.D.M.); (A.D.S.); (A.C.S.); (C.J.M.); (T.M.); (J.S.)
| | - Aaron D. Schimmer
- Princess Margaret Cancer Centre, Department of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C1, Canada; (R.C.); (A.D.L.); (S.K.); (D.M.); (S.M.C.); (V.G.); (K.W.Y.); (M.D.M.); (A.D.S.); (A.C.S.); (C.J.M.); (T.M.); (J.S.)
| | - Andre C. Schuh
- Princess Margaret Cancer Centre, Department of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C1, Canada; (R.C.); (A.D.L.); (S.K.); (D.M.); (S.M.C.); (V.G.); (K.W.Y.); (M.D.M.); (A.D.S.); (A.C.S.); (C.J.M.); (T.M.); (J.S.)
| | - Caroline J. McNamara
- Princess Margaret Cancer Centre, Department of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C1, Canada; (R.C.); (A.D.L.); (S.K.); (D.M.); (S.M.C.); (V.G.); (K.W.Y.); (M.D.M.); (A.D.S.); (A.C.S.); (C.J.M.); (T.M.); (J.S.)
| | - Tracy Murphy
- Princess Margaret Cancer Centre, Department of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C1, Canada; (R.C.); (A.D.L.); (S.K.); (D.M.); (S.M.C.); (V.G.); (K.W.Y.); (M.D.M.); (A.D.S.); (A.C.S.); (C.J.M.); (T.M.); (J.S.)
| | - Anna Xu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada; (X.L.); (A.X.)
| | - Umberto Falcone
- Department of Haematology, Istituto Nazionale Tumori IRCCS Fondazione Pascale, 80131 Naples, Italy;
| | - Jack Seki
- Princess Margaret Cancer Centre, Department of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C1, Canada; (R.C.); (A.D.L.); (S.K.); (D.M.); (S.M.C.); (V.G.); (K.W.Y.); (M.D.M.); (A.D.S.); (A.C.S.); (C.J.M.); (T.M.); (J.S.)
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada; (X.L.); (A.X.)
- Department of Pharmacy, Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada
| | - Hassan Sibai
- Princess Margaret Cancer Centre, Department of Medical Oncology and Hematology, University Health Network, Toronto, ON M5G 2C1, Canada; (R.C.); (A.D.L.); (S.K.); (D.M.); (S.M.C.); (V.G.); (K.W.Y.); (M.D.M.); (A.D.S.); (A.C.S.); (C.J.M.); (T.M.); (J.S.)
| |
Collapse
|
13
|
Harnessing hyaluronic acid-based nanoparticles for combination therapy: A novel approach for suppressing systemic inflammation and to promote antitumor macrophage polarization. Carbohydr Polym 2020; 254:117291. [PMID: 33357860 DOI: 10.1016/j.carbpol.2020.117291] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/15/2020] [Accepted: 10/19/2020] [Indexed: 01/19/2023]
Abstract
Anti-inflammatory drugs such as dexamethasone (DEX) are commonly administered to cancer patients along with anticancer drugs, however, the effect of DEX on human cancers is poorly understood. In this article, we have tailored self-assembled nanoparticles derived from hyaluronic acid (HA) wherein, anti-inflammatory DEX was used as a hydrophobic moiety for inducing amphiphilicity. The HA-DEX micelles were subsequently loaded with chemotherapeutic agent, doxorubicin (DOX) (HA-DEX-DOX) and was utilized to deliver drug cargo to human cancer cells expressing different levels of CD44 receptors. We found that DEX suppressed the cytotoxicity of DOX in HCT116, while it synergistically enhanced cytotoxicity in MCF-7 cells. When we tested DOX and HA-DEX-DOX in an ex-vivo human whole blood, we found activation of complement and the coagulation cascade in one group of donors. Encapsulation of DOX within the nanoparticle core eliminated such deleterious side-effects. The HA-DEX-DOX also polarized bone-marrow-derived anti-inflammatory M2 macrophages, to pro-inflammatory M1 phenotype with the upregulation of the cytokines TNF-α, iNOS and IL-1β.
Collapse
|
14
|
Olorundare O, Adeneye A, Akinsola A, Kolo P, Agede O, Soyemi S, Mgbehoma A, Okoye I, Albrecht R, Mukhtar H. Irvingia gabonensis Seed Extract: An Effective Attenuator of Doxorubicin-Mediated Cardiotoxicity in Wistar Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1602816. [PMID: 33149803 PMCID: PMC7603620 DOI: 10.1155/2020/1602816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/08/2020] [Accepted: 10/05/2020] [Indexed: 01/16/2023]
Abstract
Cardiotoxicity as an off-target effect of doxorubicin therapy is a major limiting factor for its clinical use as a choice cytotoxic agent. Seeds of Irvingia gabonensis have been reported to possess both nutritional and medicinal values which include antidiabetic, weight losing, antihyperlipidemic, and antioxidative effects. Protective effects of Irvingia gabonensis ethanol seed extract (IGESE) was investigated in doxorubicin (DOX)-mediated cardiotoxicity induced with single intraperitoneal injection of 15 mg/kg of DOX following the oral pretreatments of Wistar rats with 100-400 mg/kg/day of IGESE for 10 days, using serum cardiac enzyme markers (cardiac troponin I (cTI) and lactate dehydrogenase (LDH)), cardiac tissue oxidative stress markers (catalase (CAT), malonyldialdehyde (MDA), superoxide dismutase (SOD), glutathione-S-transferase (GST), glutathione peroxidase (GSH-Px), and reduced glutathione (GSH)), and cardiac histopathology endpoints. In addition, both qualitative and quantitative analyses to determine IGESE's secondary metabolites profile and its in vitro antioxidant activities were also conducted. Results revealed that serum cTnI and LDH were significantly elevated by the DOX treatment. Similarly, activities of tissue SOD, CAT, GST, and GSH levels were profoundly reduced, while GPx activity and MDA levels were profoundly increased by DOX treatment. These biochemical changes were associated with microthrombi formation in the DOX-treated cardiac tissues on histological examination. However, oral pretreatments with 100-400 mg/kg/day of IGESE dissolved in 5% DMSO in distilled water significantly attenuated increases in the serum cTnI and LDH, prevented significant alterations in the serum lipid profile and the tissue activities and levels of oxidative stress markers while improving cardiovascular disease risk indices and DOX-induced histopathological lesions. The in vitro antioxidant studies showed IGESE to have good antioxidant profile and contained 56 major secondary metabolites prominent among which are γ-sitosterol, Phytol, neophytadiene, stigmasterol, vitamin E, hexadecanoic acid and its ethyl ester, Phytyl palmitate, campesterol, lupeol, and squalene. Overall, both the in vitro and in vivo findings indicate that IGESE may be a promising prophylactic cardioprotective agent against DOX-induced cardiotoxicity, at least in part mediated via IGESE's antioxidant and free radical scavenging and antithrombotic mechanisms.
Collapse
Affiliation(s)
- Olufunke Olorundare
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Adejuwon Adeneye
- Department of Pharmacology, Therapeutics and Toxicology, Faculty of Basic Clinical Sciences, Lagos State University College of Medicine, 1-5 Oba Akinjobi Way, G.R.A., Ikeja, Lagos State, Nigeria
| | - Akinyele Akinsola
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Phillip Kolo
- Department of Medicine, Faculty of Clinical, College of Health Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Olalekan Agede
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Sunday Soyemi
- Department of Pathology and Forensic Medicine, Faculty of Basic Clinical Sciences, Lagos State University College of Medicine, 1-5 Oba Akinjobi Way, G.R.A., Ikeja, Lagos State, Nigeria
| | - Alban Mgbehoma
- Department of Pathology and Forensic Medicine, Lagos State University Teaching Hospital, 1-5 Oba Akinjobi Way, G.R.A., Ikeja, Lagos State, Nigeria
| | - Ikechukwu Okoye
- Department of Oral Pathology and Medicine, Faculty of Dentistry, Lagos State University College of Medicine, 1-5 Oba Akinjobi Way, G.R.A., Ikeja, Lagos State, Nigeria
| | - Ralph Albrecht
- Department of Animal Sciences, 1675 Observatory Drive, University of Wisconsin, Madison, WI 53706, USA
| | - Hasan Mukhtar
- Department of Dermatology, University of Wisconsin, Madison, Medical Science Center, 1300 University Avenue, Madison, WI 53706, USA
| |
Collapse
|
15
|
Lv H, Tan R, Liao J, Hao Z, Yang X, Liu Y, Xia Y. Doxorubicin contributes to thrombus formation and vascular injury by interfering with platelet function. Am J Physiol Heart Circ Physiol 2020; 319:H133-H143. [PMID: 32469636 DOI: 10.1152/ajpheart.00456.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In clinical studies, platelet aggregation and risk of thrombosis are increased in patients after doxorubicin treatment. However, the exact role of doxorubicin in platelet functions and thrombus formation in vivo remain unclear. The present study is to investigate the role of doxorubicin in platelet function in relation to thrombus formation and vascular toxicity, as well as the efficacy of antiplatelet therapy. Mice were treated with doxorubicin or vehicle (5 mg/kg iv, 4 wk), and the following parameters were determined: platelet count and size, platelet surface adhesive receptors by flow cytometry, density of granules by electron microscopy, platelet aggregation and degranulation at resting or agonist-stimulated state, platelet adhesion on fibrinogen or endothelial cells, and thrombus formation on collagen matrix. The efficacy of clopidogrel (15 mg·kg-1·day-1, followed by 5 mg·kg-1·day-1) on doxorubicin-induced changes in the aforementioned parameters as well as vascular injury were also determined. Whereas platelet count and size were similar between doxorubicin-treated and vehicle-treated mice, doxorubicin promoted thrombus formation evidenced by greater platelet aggregation, degranulation, and adhesion to endothelial cells evoked by agonists. Clopidogrel treatment attenuated the enhanced platelet activity and thrombus formation by doxorubicin, as well as vascular platelet infiltration and reactive oxygen species generation. Collectively, this study demonstrates that platelet functions are enhanced after long-term doxorubicin administration, which leads to thrombus formation and vascular toxicity, and that doxorubicin-induced changes in the functionality of platelets can be effectively inhibited by antiplatelet drugs.NEW & NOTEWORTHY Doxorubicin therapy in mice (antitumor dosage) markedly enhanced platelet functions measured as agonist-induced platelet aggregation, degranulation, and adhesion to endothelial cells, actions leading to thrombus formation and thrombosis-independent vascular injury. Clopidogrel treatment ameliorated thrombus formation and vascular toxicity induced by doxorubicin via inhibiting platelet activity.
Collapse
Affiliation(s)
- Haichen Lv
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ruopeng Tan
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiawei Liao
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhujing Hao
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaolei Yang
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yang Liu
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yunlong Xia
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
16
|
(-)-Kusunokinin inhibits breast cancer in N-nitrosomethylurea-induced mammary tumor rats. Eur J Pharmacol 2020; 882:173311. [PMID: 32619673 DOI: 10.1016/j.ejphar.2020.173311] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/17/2020] [Accepted: 06/23/2020] [Indexed: 01/10/2023]
Abstract
Natural and synthetic (-)-kusunokinin inhibited breast cancer, colon cancer and cholangiocarcinoma cells at the G2/M phase and induced apoptosis. However, there is no report on the action and adverse effects of (-)-kusunokinin in animal models. In this study, we investigated the cytotoxic effect of (-)-kusunokinin from Piper nigrum on cancer cells. NMU-induced rat mammary tumors, an ER positive breast cancer model, were treated with (-)-kusunokinin. Proteins of interest related to cell cycle, angiogenesis, migration and signaling proteins were detected in tumor tissues. Results showed that (-)-kusunokinin exhibited strong cytotoxicity against breast, colon and lung cancer cells and caused low toxicity against normal fibroblast cells. For in vivo study, 7.0 mg/kg and 14.0 mg/kg of (-)-kusunokinin reduced tumor growth without side effects on body weight, internal organs and bone marrow. Combination of (-)-kusunokinin with a low effective dose of doxorubicin significantly inhibited tumor growth and provoked cell death in cancer tissues. Mechanistically, 14.0 mg/kg of (-)-kusunokinin decreased cell proliferation (c-Src, PI3K, Akt, p-Erk1/2 and c-Myc), cell cycle (E2f-1, cyclin B1 and CDK1), and metastasis (E-cadherin, MMP-2 and MMP-9) proteins in tumor tissues, which supports its anticancer effect. We further confirmed the antimigration effect of (-)-kusunokinin; the results show that this compound inhibited breast cancer cell (MCF-7) migration in a dose-dependent manner. In conclusion, the results suggest that 14 mg/kg of (-)-kusunokinin inhibited tumors through the reduction of signaling proteins and their downstream molecules. Therefore, (-)-kusunokinin becomes an intriguing candidate for cancer treatment as it provides a strong potency in cancer inhibition.
Collapse
|
17
|
Basisty N, Kale A, Patel S, Campisi J, Schilling B. The power of proteomics to monitor senescence-associated secretory phenotypes and beyond: toward clinical applications. Expert Rev Proteomics 2020; 17:297-308. [PMID: 32425074 DOI: 10.1080/14789450.2020.1766976] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Cellular senescence is a rapidly growing field with potential relevance for the treatment of multiple human diseases. In the last decade, cellular senescence and the senescence-associated secretory phenotype (SASP) have emerged as central drivers of aging and many chronic diseases, including cancer, neurodegeneration, heart disease and osteoarthritis. Major efforts are underway to develop drugs that selectively eliminate senescent cells (senolytics) or alter the SASP (senomorphics) to treat age-related diseases in humans. The translation of senescence-targeting therapies into humans is still in early stages. Nonetheless, it is clear that proteomic approaches will facilitate the discovery of important SASP proteins, development of senescence- and SASP-derived biomarkers, and identification of therapeutic targets for senolytic and senomorphic drugs. AREAS COVERED We review recent proteomic studies of cellular senescence and their translational relevance and, particularly, characterization of the secretory phenotype and preclinical development of biomarkers (from 2008-2020, PubMed). We focus on emerging areas, such as the heterogeneity of senescent cells and the SASP, extracellular vesicles released by senescent cells, and validating biomarkers of aging in vivo. EXPERT OPINION Proteomic and multi-omic approaches will be important for the development of senescence-based biomarkers to facilitate and monitor future therapeutic interventions that target senescent cells.
Collapse
Affiliation(s)
- Nathan Basisty
- Buck Institute for Research on Aging, Novato , California, USA
| | - Abhijit Kale
- Buck Institute for Research on Aging, Novato , California, USA
| | - Sandip Patel
- Buck Institute for Research on Aging, Novato , California, USA
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato , California, USA.,Lawrence Berkeley National Laboratory, University of California , Berkeley, USA
| | | |
Collapse
|
18
|
Venturini W, Olate-Briones A, Valenzuela C, Méndez D, Fuentes E, Cayo A, Mancilla D, Segovia R, Brown NE, Moore-Carrasco R. Platelet Activation Is Triggered by Factors Secreted by Senescent Endothelial HMEC-1 Cells In Vitro. Int J Mol Sci 2020; 21:ijms21093287. [PMID: 32384773 PMCID: PMC7246568 DOI: 10.3390/ijms21093287] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/28/2020] [Accepted: 05/01/2020] [Indexed: 12/11/2022] Open
Abstract
Aging is one of the main risk factors for the development of chronic diseases, with both the vascular endothelium and platelets becoming functionally altered. Cellular senescence is a form of permanent cell cycle arrest initially described in primary cells propagated in vitro, although it can also be induced by anticancer drugs and other stressful stimuli. Attesting for the complexity of the senescent phenotype, senescent cells synthesize and secrete a wide variety of bioactive molecules. This “senescence-associated secretory phenotype” (SASP) endows senescent cells with the ability to modify the tissue microenvironment in ways that may be relevant to the development of various physiological and pathological processes. So far, however, the direct role of factors secreted by senescent endothelial cells on platelet function remains unknown. In the present work, we explore the effects of SASP factors derived from senescent endothelial cells on platelet function. To this end, we took advantage of a model in which immortalized endothelial cells (HMEC-1) were induced to senesce following exposure to doxorubicin, a chemotherapeutic drug widely used in the clinic. Our results indicate that (1) low concentrations of doxorubicin induce senescence in HMEC-1 cells; (2) senescent HMEC-1 cells upregulate the expression of selected components of the SASP and (3) the media conditioned by senescent endothelial cells are capable of inducing platelet activation and aggregation. These results suggest that factors secreted by senescent endothelial cells in vivo could have a relevant role in the platelet activation observed in the elderly or in patients undergoing therapeutic stress.
Collapse
Affiliation(s)
- Whitney Venturini
- Center for Medical Research, University of Talca Medical School, Talca 3460000, Chile; (W.V.); (A.O.-B.); (C.V.); (A.C.); (D.M.); (R.S.)
- Faculty of Health Sciences, University of Talca, Talca 3460000, Chile; (D.M.); (E.F.)
| | - Alexandra Olate-Briones
- Center for Medical Research, University of Talca Medical School, Talca 3460000, Chile; (W.V.); (A.O.-B.); (C.V.); (A.C.); (D.M.); (R.S.)
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 7500000, Chile
| | - Claudio Valenzuela
- Center for Medical Research, University of Talca Medical School, Talca 3460000, Chile; (W.V.); (A.O.-B.); (C.V.); (A.C.); (D.M.); (R.S.)
- Núcleo Científico Multidisciplinario, Universidad de Talca, Talca 3460000, Chile
| | - Diego Méndez
- Faculty of Health Sciences, University of Talca, Talca 3460000, Chile; (D.M.); (E.F.)
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, University of Talca, Talca 3460000 Chile
| | - Eduardo Fuentes
- Faculty of Health Sciences, University of Talca, Talca 3460000, Chile; (D.M.); (E.F.)
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, University of Talca, Talca 3460000 Chile
| | - Angel Cayo
- Center for Medical Research, University of Talca Medical School, Talca 3460000, Chile; (W.V.); (A.O.-B.); (C.V.); (A.C.); (D.M.); (R.S.)
- Faculty of Health Sciences, University of Talca, Talca 3460000, Chile; (D.M.); (E.F.)
| | - Daniel Mancilla
- Center for Medical Research, University of Talca Medical School, Talca 3460000, Chile; (W.V.); (A.O.-B.); (C.V.); (A.C.); (D.M.); (R.S.)
| | - Raul Segovia
- Center for Medical Research, University of Talca Medical School, Talca 3460000, Chile; (W.V.); (A.O.-B.); (C.V.); (A.C.); (D.M.); (R.S.)
- Faculty of Health Sciences, University of Talca, Talca 3460000, Chile; (D.M.); (E.F.)
| | - Nelson E. Brown
- Center for Medical Research, University of Talca Medical School, Talca 3460000, Chile; (W.V.); (A.O.-B.); (C.V.); (A.C.); (D.M.); (R.S.)
- Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Talca 3460000, Chile
- Correspondence: (N.E.B.); (R.M.-C.)
| | - Rodrigo Moore-Carrasco
- Faculty of Health Sciences, University of Talca, Talca 3460000, Chile; (D.M.); (E.F.)
- Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Talca 3460000, Chile
- Correspondence: (N.E.B.); (R.M.-C.)
| |
Collapse
|
19
|
Wei H, Harper MT. Comparison of putative BH3 mimetics AT-101, HA14-1, sabutoclax and TW-37 with ABT-737 in platelets. Platelets 2020; 32:105-112. [PMID: 32079453 DOI: 10.1080/09537104.2020.1724276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Platelet lifespan is regulated by intrinsic apoptosis. Platelet apoptosis can be triggered by BH3 mimetics that inhibit the pro-survival Bcl-2 family protein, Bcl-xL. Here, we investigated several small molecules that are reported to act as BH3 mimetics and compared their effects to the well-established BH3 mimetic, ABT-737. Platelet phosphatidylserine (PS) exposure was determined by flow cytometry. Changes in cytosolic Ca2+ signaling were detected using Cal-520. Plasma membrane integrity was determined by calcein leakage. The roles of caspases and calpain in these processes were determined using Q-VD-OPh and calpeptin, respectively. As previously reported, ABT-737 triggered PS exposure in a caspase-dependent manner and calcein loss in a caspase and calpain-dependent manner. In contrast, AT-101 and sabutoclax triggered PS exposure independently of caspases. HA14-1 also triggered PS exposure in a caspase-independent but calpain-dependent manner. There were also significant differences in the pattern and protease-dependency of cytosolic Ca2+ signaling in response to these drugs compared to ABT-737. Since there are clear differences between the action of ABT-737 and the other putative BH3 mimetics investigated here, AT-101, HA14-1 and sabutoclax cannot be considered as acting as BH3 mimetics in platelets. Furthermore, the platelet death caused by these drugs is likely to be distinct from apoptosis.
Collapse
Affiliation(s)
- Hao Wei
- Department of Pharmacology, University of Cambridge , Cambridge, UK
| | - Matthew T Harper
- Department of Pharmacology, University of Cambridge , Cambridge, UK
| |
Collapse
|
20
|
Vincristine induces procoagulant activity of the human lymphoblastic leukemia cell line Jurkat through the release of extracellular vesicles. J Thromb Thrombolysis 2020; 48:195-202. [PMID: 31175530 DOI: 10.1007/s11239-019-01894-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Thromboembolic events are frequent and serious complications of acute lymphoblastic leukaemia treatment. The importance of chemotherapy in the pathogenesis of this increased risk is enhanced by the fact that thrombosis rarely occurs at diagnosis. Our study aims at investigating the effect of chemotherapy on pro-coagulant activity (PCA), phosphatidylserine (PS) exposure, tissue factor (TF) activity and derived extracellular vesicles (EV) of Jurkat cells. Jurkat cells were treated with two commonly used chemotherapeutics: Vincristine (VCR) or Daunorubicin (DNR), at relevant concentrations. PCA of cells and derived EV were evaluated using Thrombin generation Assay (TGA). Cells or EV were incubated with annexin V or anti TF antibodies to assess the respective contribution of TF and PS. PS exposure on cells was analysed by flow cytometry. Derived EV were evaluated in fluorescence microscopy and flow cytometry. Untreated Jurkat cells and EV support thrombin generation. Thrombin generation was abolished when PS activity was inhibited by annexin V. VCR treatment resulted in a time dependent increase of thrombin generation. After VCR exposure, TF activity increased as well as PS exposure increased on the cell surface. The increase in TF activity was abolished by annexin V indicating that PS was required. A spontaneous release of EV from Jurkat cells was observed and VCR treatment increased the number of generated EV. Our results indicate that VCR increased the PCA of Jurkat cells predominantly through PS exposure and increased EV generation. Lymphoid blasts derived EV could be biomarkers to determine high thrombotic risk ALL patients.
Collapse
|
21
|
Wiley CD, Liu S, Limbad C, Zawadzka AM, Beck J, Demaria M, Artwood R, Alimirah F, Lopez-Dominguez JA, Kuehnemann C, Danielson SR, Basisty N, Kasler HG, Oron TR, Desprez PY, Mooney SD, Gibson BW, Schilling B, Campisi J, Kapahi P. SILAC Analysis Reveals Increased Secretion of Hemostasis-Related Factors by Senescent Cells. Cell Rep 2019; 28:3329-3337.e5. [PMID: 31553904 PMCID: PMC6907691 DOI: 10.1016/j.celrep.2019.08.049] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/26/2019] [Accepted: 08/14/2019] [Indexed: 01/07/2023] Open
Abstract
Cellular senescence irreversibly arrests cell proliferation, accompanied by a multi-component senescence-associated secretory phenotype (SASP) that participates in several age-related diseases. Using stable isotope labeling with amino acids (SILACs) and cultured cells, we identify 343 SASP proteins that senescent human fibroblasts secrete at 2-fold or higher levels compared with quiescent cell counterparts. Bioinformatic analysis reveals that 44 of these proteins participate in hemostasis, a process not previously linked with cellular senescence. We validated the expression of some of these SASP factors in cultured cells and in vivo. Mice treated with the chemotherapeutic agent doxorubicin, which induces widespread cellular senescence in vivo, show increased blood clotting. Conversely, selective removal of senescent cells using transgenic p16-3MR mice showed that clearing senescent cells attenuates the increased clotting caused by doxorubicin. Our study provides an in-depth, unbiased analysis of the SASP and unveils a function for cellular senescence in hemostasis.
Collapse
Affiliation(s)
| | - Su Liu
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | | | - Jennifer Beck
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | - Marco Demaria
- European Institute for the Biology of Aging, University of Groningen, Groningen, the Netherlands
| | - Robert Artwood
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | | | | | | | - Natan Basisty
- Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | | | - Pierre-Yves Desprez
- Buck Institute for Research on Aging, Novato, CA 94945, USA; California Pacific Medical Center, Research Institute, San Francisco, CA 94107, USA
| | - Sean D Mooney
- Department of Biomedical Informatics & Medical Education, University of Washington, Seattle, WA 98195, USA
| | - Bradford W Gibson
- Discovery Attribute Sciences, Amgen Inc., South San Francisco, CA 94080, USA
| | | | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA 94945, USA; Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| | - Pankaj Kapahi
- Buck Institute for Research on Aging, Novato, CA 94945, USA.
| |
Collapse
|
22
|
Fuentes E, Araya-Maturana R, Urra FA. Regulation of mitochondrial function as a promising target in platelet activation-related diseases. Free Radic Biol Med 2019; 136:172-182. [PMID: 30625393 DOI: 10.1016/j.freeradbiomed.2019.01.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/22/2018] [Accepted: 01/04/2019] [Indexed: 12/13/2022]
Abstract
Platelets are anucleated cell elements produced by fragmentation of the cytoplasm of megakaryocytes and have a unique metabolic phenotype compared with circulating leukocytes, exhibiting a high coupling efficiency to mitochondrial adenosine triphosphate production with reduced respiratory reserve capacity. Platelet mitochondria are well suited for ex vivo analysis of different diseases. Even some diseases induce mitochondrial changes in platelets without reflecting them in other organs. During platelet activation, an integrated participation of glycolysis and oxidative phosphorylation is mediated by oxidative stress production-dependent signaling. The platelet activation-dependent procoagulant activity mediated by collagen, thrombin and hyperglycemia induce mitochondrial dysfunction to promote thrombosis in oxidative stress-associated pathological conditions. Interestingly, some compounds exhibit a protective action on platelet mitochondrial dysfunction through control of mitochondrial oxidative stress production or inhibition of respiratory complexes. They can be grouped in a) Natural source-derived compounds (e.g. Xanthohumol, Salvianoloc acid A and Sila-amide derivatives of NAC), b) TPP+-linked small molecules (e.g. mitoTEMPO and mitoQuinone) and c) FDA-approved drugs (e.g. metformin and statins), illustrating the wide range of molecular structures capable of effectively interacting with platelet mitochondria. The present review article aims to discuss the mechanisms of mitochondrial dysfunction and their association with platelet activation-related diseases.
Collapse
Affiliation(s)
- Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Excellence Research Program on Healthy Aging (PIEI-ES), Universidad de Talca, Talca, Chile.
| | - Ramiro Araya-Maturana
- Instituto de Química de Recursos Naturales, Programa de Investigación Asociativa en Cáncer Gástrico (PIA-CG), Universidad de Talca, Talca, Chile
| | - Félix A Urra
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
| |
Collapse
|
23
|
Thrombin Generation and Cancer: Contributors and Consequences. Cancers (Basel) 2019; 11:cancers11010100. [PMID: 30654498 PMCID: PMC6356447 DOI: 10.3390/cancers11010100] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/04/2019] [Accepted: 01/08/2019] [Indexed: 12/19/2022] Open
Abstract
The high occurrence of cancer-associated thrombosis is associated with elevated thrombin generation. Tumour cells increase the potential for thrombin generation both directly, through the expression and release of procoagulant factors, and indirectly, through signals that activate other cell types (including platelets, leukocytes and erythrocytes). Furthermore, cancer treatments can worsen these effects. Coagulation factors, including tissue factor, and inhibitors of coagulation are altered and extracellular vesicles (EVs), which can promote and support thrombin generation, are released by tumour and other cells. Some phosphatidylserine-expressing platelet subsets and platelet-derived EVs provide the surface required for the assembly of coagulation factors essential for thrombin generation in vivo. This review will explore the causes of increased thrombin production in cancer, and the availability and utility of tests and biomarkers. Increased thrombin production not only increases blood coagulation, but also promotes tumour growth and metastasis and as a consequence, thrombin and its contributors present opportunities for treatment of cancer-associated thrombosis and cancer itself.
Collapse
|
24
|
Cytotoxicity of Air Pollutant 9,10-Phenanthrenequinone: Role of Reactive Oxygen Species and Redox Signaling. BIOMED RESEARCH INTERNATIONAL 2018; 2018:9523968. [PMID: 29984252 PMCID: PMC6015725 DOI: 10.1155/2018/9523968] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 04/30/2018] [Indexed: 01/22/2023]
Abstract
Atmospheric pollution has been a principal topic recently in the scientific and political community due to its role and impact on human and ecological health. 9,10-phenanthrenequinone (9,10-PQ) is a quinone molecule found in air pollution abundantly in the diesel exhaust particles (DEP). This compound has studied extensively and has been shown to develop cytotoxic effects both in vitro and in vivo. 9, 10-PQ has been proposed to play a critical role in the development of cytotoxicity via generation of reactive oxygen species (ROS) through redox cycling. This compound also reduces expression of glutathione (GSH), which is critical in Phase II detoxification reactions. Understanding the underlying cellular mechanisms involved in cytotoxicity can allow for the development of therapeutics designed to target specific molecules significantly involved in the 9,10-PQ-induced ROS toxicity. This review highlights the developments in the understanding of the cytotoxic effects of 9, 10-PQ with special emphasis on the possible mechanisms involved.
Collapse
|
25
|
Howden EJ, La Gerche A, Arthur JF, McMullen JR, Jennings GL, Dunstan DW, Owen N, Avery S, Kingwell BA. Standing up to the cardiometabolic consequences of hematological cancers. Blood Rev 2018; 32:349-360. [PMID: 29496356 DOI: 10.1016/j.blre.2018.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/06/2017] [Accepted: 02/20/2018] [Indexed: 12/18/2022]
Abstract
Hematological cancer survivors are highly vulnerable to cardiometabolic complications impacting long-term health status, quality of life and survival. Elevated risk of diabetes and cardiovascular disease arises not only from the effects of the cancers themselves, but also from the toxic effects of cancer therapies, and deconditioning arising from reduced physical activity levels. Regular physical activity can circumvent or reverse adverse effects on the heart, skeletal muscle, vasculature and blood cells, through a combination of systemic and molecular mechanisms. We review the link between hematological cancers and cardiometabolic risk with a focus on adult survivors, including the contributing mechanisms and discuss the potential for physical activity interventions, which may act to oppose the negative effects of both physical deconditioning and therapies (conventional and targeted) on metabolic and growth signaling (kinase) pathways in the heart and beyond. In this context, we focus particularly on strategies targeting reducing and breaking up sedentary time and provide recommendations for future research.
Collapse
Affiliation(s)
- Erin J Howden
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia.
| | - André La Gerche
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia.
| | - Jane F Arthur
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia
| | - Julie R McMullen
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia.
| | - Garry L Jennings
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia; Sydney Medical School, University of Sydney, NSW, Australia.
| | - David W Dunstan
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia.
| | - Neville Owen
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia.
| | - Sharon Avery
- Malignant Hematology and Stem Cell Transplantation Service, The Alfred Hospital, 55 Commercial Road, Melbourne, VIC, Australia.
| | - Bronwyn A Kingwell
- Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia.
| |
Collapse
|
26
|
Abstract
PURPOSE OF REVIEW Recent studies indicate that the risk of thrombosis in hematologic patients may be similar or even higher than that found in patients with solid tumors. However, available information about pathogenesis and incidence of thrombosis in acute leukemia is limited. This review focuses on mechanisms underlying thrombosis in acute leukemia and discusses recent literature data. RECENT FINDINGS In the last few years, proofs have been provided that leukemic cells release free prothrombotic products, such as micro-vesicles, tissue factors, circulating free DNA and RNA. Furthermore, leukemic blasts can activate the procoagulant population of platelets, which initiate and amplify coagulation, causing thrombosis. In addition to factors produced by acute leukemia itself, others concur to trigger thrombosis. Some drugs, infections and insertion of central venous catheter have been described to increase risk of thrombosis in patients with acute leukemia. SUMMARY Thrombosis represents a serious complication in patients affected by myeloid and lymphoid acute leukemia. A proper knowledge of its pathophysiology and of the predisposing risk factors may allow to implement strategies of prevention. Improving prevention of thrombosis appears a major goal in patients whose frequent conditions of thrombocytopenia impede an adequate delivery of anticoagulant therapy.
Collapse
|
27
|
Guan Y, Nakano D, Zhang Y, Li L, Liu W, Nishida M, Kuwabara T, Morishita A, Hitomi H, Mori K, Mukoyama M, Masaki T, Hirano K, Nishiyama A. A protease-activated receptor-1 antagonist protects against podocyte injury in a mouse model of nephropathy. J Pharmacol Sci 2017; 135:S1347-8613(17)30128-7. [PMID: 29110957 DOI: 10.1016/j.jphs.2017.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/13/2017] [Accepted: 08/15/2017] [Indexed: 12/19/2022] Open
Abstract
The kidney expresses protease-activated receptor-1 (PAR-1). PAR-1 is known as a thrombin receptor, but its role in kidney injury is not well understood. In this study, we examined the contribution of PAR-1 to kidney glomerular injury and the effects of its inhibition on development of nephropathy. Mice were divided into 3 groups: control, doxorubicin + vehicle (15 mg/kg doxorubicin and saline) and doxorubicin + Q94 (doxorubicin at 15 mg/kg and the PAR-1 antagonist Q94 at 5 mg/kg/d) groups. Where indicated, doxorubicin was administered intravenously and PAR-1 antagonist or saline vehicle by subcutaneous osmotic mini-pump. PAR-1 expression was increased in glomeruli of mice treated with doxorubicin. Q94 treatment significantly suppressed the increased albuminuria in these nephropathic mice. Pathological analysis showed that Q94 treatment significantly attenuated periodic acid-Schiff and desmin staining, indicators of podocyte injury, and also decreased glomerular levels of podocin and nephrin. Furthermore, thrombin increased intracellular calcium levels in podocytes. This increase was suppressed by Q94 and Rox4560, a transient receptor potential cation channel (TRPC)3/6 antagonist. In addition, both Q94 and Rox4560 suppressed the doxorubicin-induced increase in activities of caspase-9 and caspase-3 in podocytes. These data suggested that PAR-1 contributes to development of podocyte and glomerular injury and that PAR-1 antagonists have therapeutic potential.
Collapse
Affiliation(s)
- Yu Guan
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| | - Daisuke Nakano
- Department of Pharmacology, Kagawa University, Kagawa, Japan.
| | - Yifan Zhang
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| | - Lei Li
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| | - Wenhua Liu
- Department of Cardiovascular Physiology, Kagawa University, Kagawa, Japan
| | - Motohiro Nishida
- Division of Cardiocirculatory Signaling, Okazaki Institute for Integrative Bioscience (National Institute for Physiological Sciences), National Institutes of Natural Sciences, Okazaki, Aichi, Japan
| | - Takashige Kuwabara
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Asahiro Morishita
- Department of Gastroenterology & Neurology, Kagawa University, Kagawa, Japan
| | - Hirofumi Hitomi
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| | - Kiyoshi Mori
- Department of Nephrology and Kidney Research, Shizuoka General Hospital, Shizuoka, Japan
| | - Masashi Mukoyama
- Department of Nephrology, Kumamoto University Graduate School of Medical Sciences, Kumamoto, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology & Neurology, Kagawa University, Kagawa, Japan
| | - Katsuya Hirano
- Department of Cardiovascular Physiology, Kagawa University, Kagawa, Japan
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University, Kagawa, Japan
| |
Collapse
|
28
|
Boussada M, Ali RB, Said AB, Bokri K, Akacha AB, Dziri C, El May MV. Selenium and a newly synthesized Thiocyanoacetamide reduce Doxorubicin gonadotoxicity in male rat. Biomed Pharmacother 2017; 89:1005-1017. [DOI: 10.1016/j.biopha.2017.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/26/2017] [Accepted: 03/01/2017] [Indexed: 12/29/2022] Open
|
29
|
Gurav D, Varghese OP, Hamad OA, Nilsson B, Hilborn J, Oommen OP. Chondroitin sulfate coated gold nanoparticles: a new strategy to resolve multidrug resistance and thromboinflammation. Chem Commun (Camb) 2016; 52:966-9. [PMID: 26587574 DOI: 10.1039/c5cc09215a] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
We have developed the first chondroitin sulfate polymer coated gold nanoparticles that can simultaneously overcome mulidrug resistance in cancer cells and suppress thromboinflammation triggered by the chemotherapeutic drug.
Collapse
Affiliation(s)
- Deepanjali Gurav
- Department of Chemistry, Ångström Laboratory, Uppsala University, SE-75121, Sweden. and Department of Chemistry, Savitri Bai Phule Pune University, Maharashtra, India
| | - Oommen P Varghese
- Department of Chemistry, Ångström Laboratory, Uppsala University, SE-75121, Sweden.
| | - Osama A Hamad
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, SE-75105, Sweden
| | - Bo Nilsson
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, SE-75105, Sweden
| | - Jöns Hilborn
- Department of Chemistry, Ångström Laboratory, Uppsala University, SE-75121, Sweden.
| | - Oommen P Oommen
- Department of Chemistry, Ångström Laboratory, Uppsala University, SE-75121, Sweden.
| |
Collapse
|
30
|
Dobrovolskaia MA, Shurin M, Shvedova AA. Current understanding of interactions between nanoparticles and the immune system. Toxicol Appl Pharmacol 2016; 299:78-89. [PMID: 26739622 PMCID: PMC4811709 DOI: 10.1016/j.taap.2015.12.022] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 12/24/2015] [Accepted: 12/26/2015] [Indexed: 10/22/2022]
Abstract
The delivery of drugs, antigens, and imaging agents benefits from using nanotechnology-based carriers. The successful translation of nanoformulations to the clinic involves thorough assessment of their safety profiles, which, among other end-points, includes evaluation of immunotoxicity. The past decade of research focusing on nanoparticle interaction with the immune system has been fruitful in terms of understanding the basics of nanoparticle immunocompatibility, developing a bioanalytical infrastructure to screen for nanoparticle-mediated immune reactions, beginning to uncover the mechanisms of nanoparticle immunotoxicity, and utilizing current knowledge about the structure-activity relationship between nanoparticles' physicochemical properties and their effects on the immune system to guide safe drug delivery. In the present review, we focus on the most prominent pieces of the nanoparticle-immune system puzzle and discuss the achievements, disappointments, and lessons learned over the past 15years of research on the immunotoxicity of engineered nanomaterials.
Collapse
Affiliation(s)
- Marina A Dobrovolskaia
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Leidos Biomedical Research Inc., Frederick National Laboratory for Cancer Research, NCI at Frederick, Frederick, MD 21702, USA.
| | - Michael Shurin
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA; Department of Immunology, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
| | - Anna A Shvedova
- Health Effects Laboratory Division, National Institute of Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV 26505, USA; Department of Physiology and Pharmacology, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
31
|
Kim JH, Jeon YS, Cho SG. Successful management of four unusual cases of acute aortic thrombus induced by chemotherapy. Clin Imaging 2016; 40:224-7. [PMID: 26995575 DOI: 10.1016/j.clinimag.2015.10.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 10/06/2015] [Accepted: 10/30/2015] [Indexed: 11/30/2022]
Abstract
An acute aortic thrombus is an unusual finding, and this is a source of distal arterial embolism, which has a poor prognosis. Chemotherapeutics have been reported as possible rare causes of acute arterial thrombus. We report four cases of acute aortic thrombus after chemotherapy, which were effectively treated with systemic anticoagulation or endovascular management.
Collapse
Affiliation(s)
- Jun Ho Kim
- Department of Radiology, Inha University Hospital, Inha University School of Medicine, Inhang-ro 27, Jung-gu, Incheon, Korea 400-711
| | - Yong Sun Jeon
- Department of Radiology, Inha University Hospital, Inha University School of Medicine, Inhang-ro 27, Jung-gu, Incheon, Korea 400-711.
| | - Soon Gu Cho
- Department of Radiology, Inha University Hospital, Inha University School of Medicine, Inhang-ro 27, Jung-gu, Incheon, Korea 400-711
| |
Collapse
|
32
|
Perez-Segura P, Zamorano-León JJ, Acosta D, Santos-Sancho JM, Modrego J, Caldés T, de la Hoya M, Díaz-Rubio E, Díaz-Millán I, de Las Heras N, Rico Zalba LA, Lahera V, Melander O, López Farré A. BRCA2 gene mutations and coagulation-associated biomarkers. Thromb Haemost 2015; 115:415-23. [PMID: 26446551 DOI: 10.1160/th15-06-0520] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 09/10/2015] [Indexed: 01/01/2023]
Abstract
Thromboembolic events are the second cause of death in cancer patients, although the mechanisms underlying this increased thromboembolic risk remain unclear. The aims of this study were to examine whether BRCA2 gene mutations may modify the circulating levels of thrombocoagulation biomarkers and whether breast cancer development may influence changes in such circulating biomarkers. The study was performed in 25 women with mutations in the BRCA2 gene (n=12 breast cancer, n=13 breast cancer-free) and in 13 BRCA2 non-mutant controls. Results revealed that plasma levels of fibrinogen gamma chain isotypes 2 and 3, haptoglobin isotypes 4 and 5, serotransferrin isotypes 3 and 4 and convertase C3/C5 isotypes 4 and 5 were significantly higher in BRCA2 mutation carriers compared to controls. However, plasma levels of vitamin D binding protein isotype 1 and alpha1-antitrypsin isotypes 2, 3 and 4 were significantly decreased in BRCA2 mutation carriers compared to controls. Plasma expression of PF4 and P-selectin was significantly higher in BRCA2 mutations carriers than in controls. BRCA2 truncated mutations conserving a binding region for RAD51 were associated with increased plasma levels of alpha1-antitrypsin isotypes 3 and 4 with respect to women showing BRCA2 mutations that loss the binding RD51 region to BRCA2. Only plasma levels of vitamin D binding protein isotypes 1 and 3 were significantly reduced and alpha 1-antitrypsin isotype 1 was increased in cancer-free BRCA2 mutation carriers compared to BRCA2 mutation carriers with breast cancer. The presence of BRCA2 mutations is associated with increased plasma levels of thrombo-coagulating-related proteins, which are independent to breast cancer development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Antonio López Farré
- Antonio López Farré, Medicine Department, School of Medicine, Plaza Ramón y Cajal. SN, Universidad Complutense de Madrid, Madrid 28040, Spain, E-mail:
| |
Collapse
|
33
|
Wang Z, Wang J, Xie R, Liu R, Lu Y. Mitochondria-derived reactive oxygen species play an important role in Doxorubicin-induced platelet apoptosis. Int J Mol Sci 2015; 16:11087-100. [PMID: 25988386 PMCID: PMC4463691 DOI: 10.3390/ijms160511087] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 05/04/2015] [Accepted: 05/11/2015] [Indexed: 11/19/2022] Open
Abstract
Doxorubicin (DOX) is an effective chemotherapeutic agent; however; its use is limited by some side effects; such as cardiotoxicity and thrombocytopenia. DOX-induced cardiotoxicity has been intensively investigated; however; DOX-induced thrombocytopenia has not been clearly elucidated. Here we show that DOX-induced mitochondria-mediated intrinsic apoptosis and glycoprotein (GP)Ibα shedding in platelets. DOX did not induce platelet activation; whereas; DOX obviously reduced adenosine diphosphate (ADP)- and thrombin-induced platelet aggregation; and impaired platelet adhesion on the von Willebrand factor (vWF) surface. In addition; we also show that DOX induced intracellular reactive oxygen species (ROS) production and mitochondrial ROS generation in a dose-dependent manner. The mitochondria-targeted ROS scavenger Mito-TEMPO blocked intracellular ROS and mitochondrial ROS generation. Furthermore; Mito-TEMPO reduced DOX-induced platelet apoptosis and GPIbα shedding. These data indicate that DOX induces platelet apoptosis; and impairs platelet function. Mitochondrial ROS play a pivotal role in DOX-induced platelet apoptosis and GPIbα shedding. Therefore; DOX-induced platelet apoptosis might contribute to DOX-triggered thrombocytopenia; and mitochondria-targeted ROS scavenger would have potential clinical utility in platelet-associated disorders involving mitochondrial oxidative damage.
Collapse
Affiliation(s)
- Zhicheng Wang
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China.
| | - Jie Wang
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China.
| | - Rufeng Xie
- Blood Engineering Laboratory, Shanghai Blood Center, Shanghai 200051, China.
| | - Ruilai Liu
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China.
| | - Yuan Lu
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200040, China.
| |
Collapse
|
34
|
Johnson L, Marks D. Treatment of Platelet Concentrates with the Mirasol Pathogen Inactivation System Modulates Platelet Oxidative Stress and NF-κB Activation. Transfus Med Hemother 2015. [PMID: 26195930 DOI: 10.1159/000403245] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Pathogen inactivation (PI) technologies for platelets aim to improve transfusion safety by preventing the replication of contaminating pathogens. However, as a consequence of treatment, aspects of the platelet storage lesion are amplified. Mirasol treatment also affects platelet signal transduction and apoptotic protein expression. The aim of this study was to examine the effect of Mirasol treatment on the generation of reactive oxygen species (ROS) and subsequent oxidative stress. METHODS Pooled platelet concentrates were prepared in platelet-additive solution (70% SSP+ / 30% plasma). ABO-matched platelets were pooled and split, and treated with the Mirasol system (TerumoBCT) or left untreated as a control. Platelet samples were tested on day 1, 5, and 7 post-collection. RESULTS Mirasol-treated platelets had increased formation of ROS by day 5 of storage. Oxidative damage, in the form of protein carbonylation, was higher in Mirasol-treated platelets, whilst no effect on nitrotyrosine formation or lipid peroxidation was detected. The NF-κB signaling pathway was also activated in Mirasol-treated platelets, with increased expression and phosphorylation of NF-κB p65 and IκBα. CONCLUSION These data demonstrate that Mirasol-treated platelets produce more ROS and display protein alterations consistent with oxidative damage.
Collapse
Affiliation(s)
- Lacey Johnson
- Research and Development, Australian Red Cross Blood Service, Sydney, NSW, Australia
| | - Denese Marks
- Research and Development, Australian Red Cross Blood Service, Sydney, NSW, Australia
| |
Collapse
|
35
|
Gordiienko IА, SE Dnipropetrovsk Medical Academy of Health Ministry of Ukraine. Activity of trypsin-like enzymes and gelatinases in rats with doxorubicin cardiomyopathy. UKRAINIAN BIOCHEMICAL JOURNAL 2014. [DOI: 10.15407/ubj86.06.139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
36
|
Pietraforte D, Vona R, Marchesi A, de Jacobis IT, Villani A, Del Principe D, Straface E. Redox control of platelet functions in physiology and pathophysiology. Antioxid Redox Signal 2014; 21:177-93. [PMID: 24597688 DOI: 10.1089/ars.2013.5532] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
SIGNIFICANCE An imbalance between the production and the detoxification of reactive oxygen species and reactive nitrogen species (ROS/RNS) can be implicated in many pathological processes. Platelets are best known as primary mediators of hemostasis and can be either targets of ROS/RNS or generate radicals during cell activation. These conditions can dramatically affect platelet physiology, leading even, as an ultimate event, to the cell number modification. In this case, pathological conditions such as thrombocytosis (promoted by increased cell number) or thrombocytopenia and myelodysplasia (promoted by cell decrease mediated by accelerated apoptosis) can occur. RECENT ADVANCES Usually, in peripheral blood, ROS/RNS production is balanced by the rate of oxidant elimination. Under this condition, platelets are in a nonadherent "resting" state. During endothelial dysfunction or under pathological conditions, ROS/RNS production increases and the platelets respond with specific biochemical and morphologic changes. Mitochondria are at the center of these processes, being able to both generate ROS/RNS, that drive redox-sensitive events, and respond to ROS/RNS-mediated changes of the cellular redox state. Irregular function of platelets and enhanced interaction with leukocytes and endothelial cells can contribute to pathogenesis of atherosclerotic and thrombotic events. CRITICAL ISSUES The relationship between oxidative stress, platelet death, and the activation-dependent pathways that drive platelet pro-coagulant activity is unclear and deserves to be explored. FUTURE DIRECTIONS Expanding knowledge about how platelets can mediate hemostasis and modulate inflammation may lead to novel and effective therapeutic strategies for the long and growing list of pathological conditions that involve both thrombosis and inflammation.
Collapse
Affiliation(s)
- Donatella Pietraforte
- 1 Department of Cell Biology and Neurosciences, Section of Cell Aging and Gender Medicine, Istituto Superiore di Sanità , Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
37
|
Kędzierska M, Malinowska J, Kontek B, Kołodziejczyk-Czepas J, Czernek U, Potemski P, Piekarski J, Jeziorski A, Olas B. Chemotherapy modulates the biological activity of breast cancer patients plasma: The protective properties of black chokeberry extract. Food Chem Toxicol 2013; 53:126-32. [DOI: 10.1016/j.fct.2012.11.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 11/23/2012] [Accepted: 11/24/2012] [Indexed: 11/26/2022]
|
38
|
Lin MC, Yin MC. Preventive Effects of Ellagic Acid Against Doxorubicin-Induced Cardio-Toxicity in Mice. Cardiovasc Toxicol 2013; 13:185-93. [DOI: 10.1007/s12012-013-9197-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
39
|
Rashidi A, Silverberg ML, Conkling PR, Fisher SI. Thrombosis in acute promyelocytic leukemia. Thromb Res 2012; 131:281-9. [PMID: 23266518 DOI: 10.1016/j.thromres.2012.11.024] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 11/20/2012] [Accepted: 11/23/2012] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Compared to bleeding, major thromboses are a less commonly encountered problem in acute promyelocytic leukemia (APL), and our knowledge about the epidemiology of major thromboses in APL stems mainly from individual case reports. The purpose of this study was to provide a better understanding of the epidemiology of APL-related thrombosis as a first step towards developing preventive strategies. MATERIALS AND METHODS We report a rare case of catastrophic acute myocardial infarction in a patient with APL while she developed the all-trans retinoic acid (ATRA) syndrome. We describe the pathogenesis of APL-related thrombosis and review all previously reported cases of major thromboses in APL. RESULTS We found 94 cases of major thromboses in patients with APL. Both genders were almost equally affected. More than 80% of events occurred before or during induction therapy with deep vein thrombosis/pulmonary embolism (DVT/PE), cardiac events, and cerebrovascular accidents (CVA) constituting more than 75% of all cases. Arterial events were slightly more common than venous events. Only 2 arterial events occurred after completion of induction therapy. Thrombosis was associated with life-threatening hemorrhage in about 15%, significant coagulative defects in about 50%, and ATRA syndrome in about 13% of cases. Cardiac thrombotic events, DVT/PE, and CVA were associated with ATRA syndrome in 24%, 4.5%, and 5% of cases, respectively (p=0.09). None of the observed trends and associations reached statistical significance. CONCLUSIONS This review advances our understanding of the epidemiology of major thromboses in APL. With accumulation of more cases in the literature, some of our results may become statistically significant.
Collapse
Affiliation(s)
- Armin Rashidi
- Department of Internal Medicine, Eastern Virginia Medical School, Norfolk, VA, USA.
| | | | | | | |
Collapse
|
40
|
Wieschhaus AJ, Le Breton GC, Chishti AH. Headpiece domain of dematin regulates calcium mobilization and signaling in platelets. J Biol Chem 2012; 287:41218-31. [PMID: 23060452 DOI: 10.1074/jbc.m112.364679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Dematin is a broadly expressed membrane cytoskeletal protein that has been well characterized in erythrocytes and to a lesser extent in non-erythroid cells. However, dematin's function in platelets is not known. Here, we show that dematin is abundantly expressed in both human and mouse platelets. Platelets harvested from the dematin headpiece knock-out (HPKO) mouse model exhibit a striking defect in the mobilization of calcium in response to multiple agonists of platelet activation. The reduced calcium mobilization in HPKO platelets is associated with concomitant inhibition of platelet aggregation and granule secretion. Integrin α(IIb)β(3) activation in response to agonists is attenuated in the HPKO platelets. The mutant platelets show nearly normal spreading on fibrinogen and an unaltered basal cAMP level; however, the clot retraction was compromised in the mutant mice. Immunofluorescence analysis indicated that dematin is present both at the dense tubular system and plasma membrane fractions of platelets. Proteomic analysis of dematin-associated proteins in human platelets identified inositol 1,4,5-trisphosphate 3-kinase isoform B (IP3KB) as a binding partner, which was confirmed by immunoprecipitation analysis. IP3KB, a dense tubular system protein, is a major regulator of calcium homeostasis. Loss of the dematin headpiece resulted in a decrease of IP3KB at the membrane and increased levels of IP3KB in the cytosol. Collectively, these findings unveil dematin as a novel regulator of internal calcium mobilization in platelets affecting multiple signaling and cytoskeletal functions. Implications of a conserved role of dematin in the regulation of calcium homeostasis in other cell types will be discussed.
Collapse
Affiliation(s)
- Adam J Wieschhaus
- Department of Molecular Physiology and Pharmacology, Tufts University School of Medicine, Boston, Massachusetts 02111, USA
| | | | | |
Collapse
|
41
|
Kedzierska M, Czernek U, Szydłowska-Pazera K, Potemski P, Piekarski J, Jeziorski A, Olas B. The changes of blood platelet activation in breast cancer patients before surgery, after surgery, and in various phases of the chemotherapy. Platelets 2012; 24:462-8. [DOI: 10.3109/09537104.2012.711866] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|