51
|
Neuronal glutamatergic changes and peripheral markers of cytoskeleton dynamics change synchronically 24 h after sub-anaesthetic dose of ketamine in healthy subjects. Behav Brain Res 2019; 359:312-319. [DOI: 10.1016/j.bbr.2018.10.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 10/15/2018] [Accepted: 10/15/2018] [Indexed: 12/20/2022]
|
52
|
Grau M, Cremer JM, Schmeichel S, Kunkel M, Bloch W. Comparisons of Blood Parameters, Red Blood Cell Deformability and Circulating Nitric Oxide Between Males and Females Considering Hormonal Contraception: A Longitudinal Gender Study. Front Physiol 2018; 9:1835. [PMID: 30618840 PMCID: PMC6305760 DOI: 10.3389/fphys.2018.01835] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 12/06/2018] [Indexed: 11/23/2022] Open
Abstract
Red blood cell (RBC) deformability is an important determinant of the microcirculation. It is influenced by various hematological parameters but also by nitric oxide (NO) which is produced in RBC from L-arginine by RBC-NO synthase. Longitudinal studies on blood profile, deformability at rest and NO levels but also differences between males and females (±hormonal contraception; HC) are less known so far. The study thus aimed to investigate RBC deformability, RBC NO species (nitrite, RxNO), RBC L-arginine concentration and basal blood parameters in males and females (±HC) as a function of time. RBC deformability was measured at rest once per week and the remaining parameters were measured once per month, respectively. A second experiment aimed to daily measure RBC deformability and 17β-estradiol in Female ± HC during a whole menstruation cycle to investigate a possible relation of the two parameters. Measured parameters showed low week-to-week variation and remained constant during study period. However, RBC deformability increased in Female + HC during study period possibly because of increasing training volume of the participants. Overall, results indicate gender differences in hematological parameters with higher RBC parameters (RBC count, hematocrit, hemoglobin concentration) in males compared to females. Differences were also observed between the female groups with Females - HC showing lower number of RBC but higher MCV and hematocrit compared to Females + HC. RBC deformability was highest in Females - HC which might be related to permanent higher estradiol levels and/or higher RBC NO levels because RBC nitrite and RBC RxNO concentrations were also highest in Females-HC. Results of the second experiment also suggest higher RBC deformability in Female - HC because of higher estradiol concentrations. L-arginine levels known to be related to RBC NO production were comparable in all groups. In conclusion, hematological, hemorheological and NO related parameters show gender differences. In particular, RBC deformability is affected by training volume and RBC estradiol concentrations. The results add new information on the complex regulation of RBC function which might help to better understand the role of RBC in the microcirculation.
Collapse
Affiliation(s)
- Marijke Grau
- Molecular and Cellular Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - John Maxwell Cremer
- Molecular and Cellular Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Steffen Schmeichel
- Molecular and Cellular Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Markus Kunkel
- Molecular and Cellular Sports Medicine, German Sport University Cologne, Cologne, Germany
| | - Wilhelm Bloch
- Molecular and Cellular Sports Medicine, German Sport University Cologne, Cologne, Germany
| |
Collapse
|
53
|
Lim HH, Jeong IH, An GD, Woo KS, Kim KH, Kim JM, Cha JK, Han JY. Early prediction of severity in acute ischemic stroke and transient ischemic attack using platelet parameters and neutrophil-to-lymphocyte ratio. J Clin Lab Anal 2018; 33:e22714. [PMID: 30411816 DOI: 10.1002/jcla.22714] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 10/15/2018] [Accepted: 10/16/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND It is still not easy to predict severity promptly in patients with acute ischemic stroke (AIS) and transient ischemic attack (TIA). We investigated that platelet parameters or combinations of them could be a useful tool for early prediction of severity of AIS and TIA at admission and after 3 months. METHODS We prospectively recruited 104 patients newly diagnosed with AIS and TIA. We investigated their neutrophil-to-lymphocyte ratio (NLR) and platelet parameters. According to the Modified Rankin Scale scores, the patients were divided into two groups. RESULTS In receiver operating characteristic (ROC) curve analyses, mean platelet volume (MPV), NLR/platelet count (PLT), MPV/PLT, MPV*NLR, and MPV*NLR/PLT showed statistically significant results in both at admission and after 3 months. Values of area under ROC curves for those tests at admission were 0.646, 0.697, 0.664, 0.708, and 0.722, respectively. Also, values after 3 months were 0.591, 0.661, 0.638, 0.662, and 0.689, respectively. CONCLUSION MPV*NLR/PLT could be used as a relatively good tool for predicting severity at the time of admission and after 3 months than other parameters or combinations of them. Further studies have to be carried out to investigate the best parameter for predicting the severity of AIS and TIA.
Collapse
Affiliation(s)
- Hyeon-Ho Lim
- Department of Laboratory Medicine, Dong-A University College of Medicine, Busan, Korea
| | - In-Hwa Jeong
- Department of Laboratory Medicine, Dong-A University College of Medicine, Busan, Korea
| | - Gyu-Dae An
- Department of Laboratory Medicine, Dong-A University College of Medicine, Busan, Korea
| | - Kwang-Sook Woo
- Department of Laboratory Medicine, Dong-A University College of Medicine, Busan, Korea
| | - Kyeong-Hee Kim
- Department of Laboratory Medicine, Dong-A University College of Medicine, Busan, Korea
| | - Jeong-Man Kim
- Department of Laboratory Medicine, Dong-A University College of Medicine, Busan, Korea
| | - Jae-Kwan Cha
- Department of Neurology, Dong-A University College of Medicine, Busan, Korea
| | - Jin-Yeong Han
- Department of Laboratory Medicine, Dong-A University College of Medicine, Busan, Korea
| |
Collapse
|
54
|
Nah EH, Kim S, Cho S, Cho HI. Complete Blood Count Reference Intervals and Patterns of Changes Across Pediatric, Adult, and Geriatric Ages in Korea. Ann Lab Med 2018; 38:503-511. [PMID: 30027692 PMCID: PMC6056383 DOI: 10.3343/alm.2018.38.6.503] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 04/05/2018] [Accepted: 06/27/2018] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Sampling a healthy reference population to generate reference intervals (RIs) for complete blood count (CBC) parameters is not common for pediatric and geriatric ages. We established age- and sex-specific RIs for CBC parameters across pediatric, adult, and geriatric ages using secondary data, evaluating patterns of changes in CBC parameters. METHODS The reference population comprised 804,623 health examinees (66,611 aged 3-17 years; 564,280 aged 18-59 years; 173,732 aged 60-99 years), and, we excluded 22,766 examinees after outlier testing. The CBC parameters (red blood cell [RBC], white blood cell [WBC], and platelet parameters) from 781,857 examinees were studied. We determined statistically significant partitions of age and sex, and calculated RIs according to the CLSI C28-A3 guidelines. RESULTS RBC parameters increased with age until adulthood and decreased with age in males, but increased before puberty and then decreased with age in females. WBC and platelet counts were the highest in early childhood and decreased with age. Sex differences in each age group were noted: WBC count was higher in males than in females during adulthood, but platelet count was higher in females than in males from puberty onwards (P<0.001). Neutrophil count was the lowest in early childhood and increased with age. Lymphocyte count decreased with age after peaking in early childhood. Eosinophil count was the highest in childhood and higher in males than in females. Monocyte count was higher in males than in females (P<0.001). CONCLUSIONS We provide comprehensive age- and sex-specific RIs for CBC parameters, which show dynamic changes with both age and sex.
Collapse
Affiliation(s)
- Eun Hee Nah
- Department of Laboratory Medicine and Health Promotion Research Institute, Korea Association of Health Promotion, Seoul, Korea.
| | - Suyoung Kim
- Department of Laboratory Medicine and Health Promotion Research Institute, Korea Association of Health Promotion, Seoul, Korea
| | - Seon Cho
- Department of Laboratory Medicine and Health Promotion Research Institute, Korea Association of Health Promotion, Seoul, Korea
| | - Han Ik Cho
- MEDIcheck LAB, Korea Association of Health Promotion, Cheongju, Korea
| |
Collapse
|
55
|
Ya F, Tian J, Li Q, Chen L, Ren J, Zhao Y, Wan J, Ling W, Yang Y. Cyanidin-3-O-β-glucoside, a Natural Polyphenol, Exerts Proapoptotic Effects on Activated Platelets and Enhances Megakaryocytic Proplatelet Formation. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:10712-10720. [PMID: 30226049 DOI: 10.1021/acs.jafc.8b03266] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
This study investigated whether the anthocyanin cyanidin-3-O-β-glucoside (Cy-3-g) could affect platelet apoptosis and proplatelet formation in vitro. Thrombin-stimulated or resting human platelets and Meg-01 megakaryocytes were incubated with Cy-3-g (0, 0.5, 5, or 50 μM). We found that the percentage of the platelet mitochondrial membrane potential treated with 5 and 50 μM Cy-3-g was significantly higher than control (15.50% ± 3.24% and 29.77% ± 4.06% versus 2.76% ± 1.33%, respectively; P < 0.05). Treatment with 5 and 50 μM Cy-3-g significantly increased phosphatidylserine exposure compared with control (40.56% ± 10.53% and 76.62% ± 8.28% versus 15.43% ± 3.93%, respectively; P < 0.05). Moreover, Cy-3-g significantly increased the expression of Bax, Bak, and cytochrome c while markedly decreasing Bcl-xL and Bcl-2 expression as well as stimulating caspase-3, caspase-9, caspase-8, Bid, and gelsolin cleavage in thrombin-activated platelets in a dose-dependent manner ( P < 0.05). However, no significant differences were observed in the apoptosis of resting platelets when treated with Cy-3-g ( P > 0.05). Furthermore, Cy-3-g significantly ( P < 0.05) enhanced cell viability (50 μM versus control, 1.34 ± 0.01 versus 0.35 ± 0.02), the number of colony-forming unit-megakaryocytes (50 μM versus control, 38 ± 3 versus 8 ± 3), CD41 expression (50 μM versus control, 96.80% ± 2.55% versus 25.57% ± 2.86%), DNA ploidy (16N) (50 μM versus control, 19.73% ± 2.34% versus 4.42% ± 1.96%), and proplatelet formation (50 μM versus control, 27.5% ± 3.77% versus 7.67% ± 2.25%) in Meg-01 cells. In conclusion, Cy-3-g promotes activated platelet apoptosis and enhances megakaryocyte proliferation, differentiation, and proplatelet formation in vitro.
Collapse
Affiliation(s)
- Fuli Ya
- Department of Nutrition, School of Public Health , Sun Yat-sen University , Guangzhou , Guangdong Province 510080 , China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou , Guangdong Province 510080 , China
- Guangdong Engineering Technology Research Center of Nutrition Translation , Guangzhou , Guangdong Province 510080 , China
| | - Jinju Tian
- Department of Pharmaceutical Analysis, School of Pharmaceutical Sciences , Sun Yat-sen University , Guangzhou , Guangdong Province 510006 , China
| | - Qing Li
- Department of Nutrition, School of Public Health , Sun Yat-sen University , Guangzhou , Guangdong Province 510080 , China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou , Guangdong Province 510080 , China
- Guangdong Engineering Technology Research Center of Nutrition Translation , Guangzhou , Guangdong Province 510080 , China
| | - Liyi Chen
- Department of Gynecology and Obstetrics, Bao'an Maternal and Child Health Hospital , Jinan University , Shenzhen 518101 , China
| | - Jing Ren
- Baoji Center For Disease Control and Prevention , Baoji , Shaanxi Province 721006 , China
| | - Yimin Zhao
- School of Public Health (Shenzhen) , Sun Yat-sen University , Guangzhou , Guangdong Province 510006 , China
| | - Jianbo Wan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences , University of Macau , Taipa , Macao 999078 , China
| | - Wenhua Ling
- Department of Nutrition, School of Public Health , Sun Yat-sen University , Guangzhou , Guangdong Province 510080 , China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou , Guangdong Province 510080 , China
- Guangdong Engineering Technology Research Center of Nutrition Translation , Guangzhou , Guangdong Province 510080 , China
| | - Yan Yang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou , Guangdong Province 510080 , China
- Guangdong Engineering Technology Research Center of Nutrition Translation , Guangzhou , Guangdong Province 510080 , China
- School of Public Health (Shenzhen) , Sun Yat-sen University , Guangzhou , Guangdong Province 510006 , China
| |
Collapse
|
56
|
Getting the Most: Enhancing Efficacy by Promoting Erythropoiesis and Thrombopoiesis after Gene Therapy in Mice with Hurler Syndrome. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 11:52-64. [PMID: 30397627 PMCID: PMC6205327 DOI: 10.1016/j.omtm.2018.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/04/2018] [Indexed: 11/24/2022]
Abstract
Novel strategies are needed to solve the conundrum of achieving clinical efficacy with high vector copy numbers (VCNs) in hematopoietic stem cells (HSCs) while attempting to minimize the potential risk of oncogenesis in lentiviral vector (LV)-mediated gene therapy clinical trials. We previously reported the benefits of reprogramming erythroid-megakaryocytic (EMK) cells for high-level lysosomal enzyme production with less risk of activating oncogenes in HSCs. Herein, using a murine model of mucopolysaccharidosis type I (MPS I) with a deficiency of α-L-iduronidase (IDUA), we sought to determine the transgene minimum effective doses (MEDs) in major organs, and if a transient increase of IDUA-containing red blood cells and platelets by repeated phlebotomy would provide further therapeutic benefits in diseased mice after EMK-restricted LV-mediated gene therapy. The MEDs for complete metabolic correction ranged from 0.1 to 2 VCNs in major visceral organs, which were dramatically reduced to 0.005–0.1 VCN by one cycle of stress induction and were associated with a further reduction of pathological deficits in mice with 0.005 VCN. This work provides a proof of concept that transiently stimulating erythropoiesis and thrombopoiesis can further improve therapeutic benefits in HSC-mediated gene therapy for MPS I, a repeatable and reversible approach to enhance clinical efficacy in the treatment of lysosomal storage diseases.
Collapse
|
57
|
Chiu CH, Higashikawa R, Yeh WL, Lei KF, Chen ACY. Investigation of Growth Factor and Tenocyte Proliferation Induced by Platelet Rich Plasma (PRP) in a 3-Chamber Co-Culture Device. MICROMACHINES 2018; 9:mi9090446. [PMID: 30424379 PMCID: PMC6187681 DOI: 10.3390/mi9090446] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/04/2018] [Accepted: 09/05/2018] [Indexed: 02/07/2023]
Abstract
The platelet-rich plasma (PRP) has become an attractive topic for soft tissue healing therapy recently. While some clinical reports revealed the effective treatments for knee osteoarthritis, lateral epicondylitis, and rotator cuff tears, other case studies showed that there was no statistically significant healing improvement. The efficacy of the PRP therapy is still unclear clinically. Thus, a significant amount of basic studies should be conducted to optimize the preparation procedure and the platelet concentration of the PRP. In this work, a 3-chamber co-culture device was developed for the PRP study in order to reduce the usage of primary cells and to avoid the PRP gelation effect. The device was a culture, well partitioning into 3 sub-chambers. Tenocytes and PRP could be respectively loaded into the sub-chambers and co-cultured under the interlinked medium. The results showed that a higher platelet number in the PRP could diffuse higher concentration of the growth factors in the medium and induce higher tenocyte proliferation. The 3-chamber co-culture device provides a simple and practical tool for the PRP study. It is potentially applied for optimizing the preparation procedure and platelet concentration of the PRP therapy.
Collapse
Affiliation(s)
- Chih-Hao Chiu
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan.
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou 333, Taiwan.
| | - Rei Higashikawa
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan.
| | - Wen-Ling Yeh
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou 333, Taiwan.
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Linkou 333, Taiwan.
| | - Kin Fong Lei
- Graduate Institute of Biomedical Engineering, Chang Gung University, Taoyuan 333, Taiwan.
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou 333, Taiwan.
| | - Alvin Chao-Yu Chen
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou 333, Taiwan.
- Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Linkou 333, Taiwan.
| |
Collapse
|
58
|
Strassel C, Gachet C, Lanza F. On the Way to in vitro Platelet Production. Front Med (Lausanne) 2018; 5:239. [PMID: 30211166 PMCID: PMC6120994 DOI: 10.3389/fmed.2018.00239] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 08/06/2018] [Indexed: 12/16/2022] Open
Abstract
The severely decreased platelet counts (10–30. 103 platelets/μL) frequently observed in patients undergoing chemotherapy, radiation treatment, or organ transplantation are associated with life-threatening increased bleeding risks. To circumvent these risks, platelet transfusion remains the treatment of choice, despite some limitations which include a limited shelf-life, storage-related deterioration, the development of alloantibodies in recipients and the transmission of infectious diseases. A sustained demand has evolved in recent years for controlled blood products, free of infectious, inflammatory, and immune risks. As a consequence, the challenge for blood centers in the near future will be to ensure an adequate supply of blood platelets, which calls for a reassessment of our transfusion models. To meet this challenge, many laboratories are now turning their research efforts toward the in vitro and customized production of blood platelets. In recent years, there has been a major enthusiasm for the cultured platelet production, as illustrated by the number of reviews that have appeared in recent years. The focus of the present review is to critically asses the arguments put forward in support of the culture of platelets for transfusion purposes. In light of this, we will recapitulate the main advances in this quickly evolving field, while noting the technical limitations to overcome to make cultured platelet a transfusional alternative.
Collapse
Affiliation(s)
- Catherine Strassel
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - Christian Gachet
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| | - François Lanza
- Université de Strasbourg, INSERM, EFS Grand Est, BPPS UMR-S 1255, FMTS, Strasbourg, France
| |
Collapse
|
59
|
Strassel C, Gachet C, Lanza F. On the way to in vitro platelet production. Transfus Clin Biol 2018; 25:220-227. [PMID: 30150135 DOI: 10.1016/j.tracli.2018.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Accepted: 07/16/2018] [Indexed: 02/07/2023]
Abstract
The severely decreased platelet counts (10-30.103 platelets/μL) frequently observed in patients undergoing chemotherapy, radiation treatment or organ transplantation are associated with life-threatening increased bleeding risks. To circumvent these risks, platelet transfusion remains the treatment of choice, despite some limitations which include a limited shelf-life, storage-related deterioration, the development of alloantibodies in recipients and the transmission of infectious diseases. A sustained demand has evolved in recent years for controlled blood products, free of infectious, inflammatory and immune risks. As a consequence, the challenge for blood centers in the near future will be to ensure an adequate supply of blood platelets, which calls for a reassessment of our transfusion models. To meet this challenge, many laboratories are now turning their research efforts towards the in vitro and customized production of blood platelets.
Collapse
Affiliation(s)
- Catherine Strassel
- Université de Strasbourg, Inserm, EFS Grand Est, BPPS UMR-S 1255, FMTS, 67000 Strasbourg, France
| | - Christian Gachet
- Université de Strasbourg, Inserm, EFS Grand Est, BPPS UMR-S 1255, FMTS, 67000 Strasbourg, France.
| | - François Lanza
- Université de Strasbourg, Inserm, EFS Grand Est, BPPS UMR-S 1255, FMTS, 67000 Strasbourg, France
| |
Collapse
|
60
|
Platelet membrane-based and tumor-associated platelettargeted drug delivery systems for cancer therapy. Front Med 2018; 12:667-677. [DOI: 10.1007/s11684-017-0583-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/24/2017] [Indexed: 12/17/2022]
|
61
|
Increased platelet count and reticulated platelets in recently symptomatic versus asymptomatic carotid artery stenosis and in cerebral microembolic signal-negative patient subgroups: results from the HaEmostasis In carotid STenosis (HEIST) study. J Neurol 2018; 265:1037-1049. [PMID: 29476243 DOI: 10.1007/s00415-018-8797-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 02/10/2018] [Accepted: 02/12/2018] [Indexed: 12/24/2022]
Abstract
BACKGROUND The pathophysiological mechanisms responsible for the disparity in stroke risk between asymptomatic and symptomatic carotid stenosis patients are not fully understood. The functionally important reticulated platelet fraction and reticulocytes could play a role. OBJECTIVES We performed a prospective, multi-centre, observational analytical study comparing full blood count parameters and platelet production/turnover/activation markers in patients with asymptomatic versus recently symptomatic moderate (≥ 50-69%) or severe (≥ 70-99%) carotid stenosis. PATIENTS/METHODS Data from 34 asymptomatic patients were compared with 43 symptomatic patients in the 'early phase' (≤ 4 weeks) and 37 of these patients in the 'late phase' (≥ 3 months) after TIA/ischaemic stroke. Reticulated platelets were quantified by whole blood flow cytometry and reticulated platelets and red cell reticulocytes by 'automated assays' (Sysmex XE-2100™). Bilateral simultaneous transcranial Doppler ultrasound monitoring classified patients as micro-embolic signal (MES)+ve or MES-ve. RESULTS Mean platelet count was higher in early (216 × 109/L; P = 0.04) and late symptomatic (219 × 109/L; P = 0.044) than asymptomatic patients (194 × 109/L). Mean platelet volume was higher in early symptomatic than asymptomatic patients (10.8 vs. 10.45 fl; P = 0.045). Automated assays revealed higher % reticulated platelet fractions in early (5.78%; P < 0.001) and late symptomatic (5.11%; P = 0.01) than asymptomatic patients (3.48%). Red cell reticulocyte counts were lower in early (0.92%; P = 0.035) and late symptomatic (0.93%; P = 0.036) than asymptomatic patients (1.07%). The automated % reticulated platelet fraction was also higher in early symptomatic than asymptomatic MES-ve patients (5.7 vs. 3.55%; P = 0.001). DISCUSSION The combination of increased platelet counts and a shift towards production of an increased population of larger, young, reticulated platelets could contribute to a higher risk of first or recurrent cerebrovascular events in recently symptomatic versus asymptomatic carotid stenosis, including those who are MES-ve.
Collapse
|
62
|
Myelodysplastic Syndrome/Acute Myeloid Leukemia Arising in Idiopathic Erythrocytosis. Case Rep Hematol 2018; 2018:4378310. [PMID: 29682367 PMCID: PMC5842720 DOI: 10.1155/2018/4378310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/07/2018] [Indexed: 11/17/2022] Open
Abstract
The term “idiopathic erythrocytosis (IE)” is applied to those cases where a causal clinical or pathological event cannot be elucidated and likely reflects a spectrum of underlying medical and molecular abnormalities. The clinical course of a patient with IE is described manifesting as a persistent erythrocytosis with a low serum erythropoietin level, mild eosinophilia, and with evidence of a thrombotic event. The patient subsequently developed a myelodysplasic syndrome (MDS) and acute myeloid leukemia (AML), an event not observed in erythrocytosis patients other than those with polycythemia vera (PV). Application of a next-generation sequencing (NGS) approach targeted for myeloid malignancies confirmed wild-type JAK2 exons 12–15 and identified a common SH2B3 W262R single-nucleotide polymorphism associated with the development of hematological features of myeloproliferative neoplasms (MPNs). Further NGS analysis detected a CBL L380P mutated clone expanding in parallel with the development of MDS and subsequent AML. Despite the absence of JAK2, MPL exon 10, or CALR exon 9 mutations, a similarity with the disease course of PV/MPN was evident. A clonal link between the erythrocytosis and AML could be neither confirmed nor excluded. Future molecular identification of the mechanisms underlying IE is likely to provide a more refined therapeutic approach.
Collapse
|
63
|
Force-activatable biosensor enables single platelet force mapping directly by fluorescence imaging. Biosens Bioelectron 2017; 100:192-200. [PMID: 28915383 DOI: 10.1016/j.bios.2017.09.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/25/2017] [Accepted: 09/05/2017] [Indexed: 11/23/2022]
Abstract
Integrin-transmitted cellular forces are critical for platelet adhesion, activation, aggregation and contraction during hemostasis and thrombosis. Measuring and mapping single platelet forces are desired in both research and clinical applications. Conventional force-to-strain based cell traction force microscopies have low resolution which is not ideal for cellular force mapping in small platelets. To enable platelet force mapping with submicron resolution, we developed a force-activatable biosensor named integrative tension sensor (ITS) which directly converts molecular tensions to fluorescent signals, therefore enabling cellular force mapping directly by fluorescence imaging. With ITS, we mapped cellular forces in single platelets at 0.4µm resolution. We found that platelet force distribution has strong polarization which is sensitive to treatment with the anti-platelet drug tirofiban, suggesting that the ITS force map can report anti-platelet drug efficacy. The ITS also calibrated integrin molecular tensions in platelets and revealed two distinct tension levels: 12-54 piconewton (nominal values) tensions generated during platelet adhesion and tensions above 54 piconewton generated during platelet contraction. Overall, the ITS is a powerful biosensor for the study of platelet mechanobiology, and holds great potential in antithrombotic drug development and assessing platelet activity in health and disease.
Collapse
|
64
|
Goldstein Z, Corneil TA, Greene DN. When Gender Identity Doesn't Equal Sex Recorded at Birth: The Role of the Laboratory in Providing Effective Healthcare to the Transgender Community. Clin Chem 2017; 63:1342-1352. [PMID: 28679645 DOI: 10.1373/clinchem.2016.258780] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 01/24/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Transgender is an umbrella term used to describe individuals who identify with a gender incongruent to or variant from their sex recorded at birth. Affirming gender identity through a variety of social, medical, and surgical interventions is critical to the mental health of transgender individuals. In recent years, awareness surrounding transgender identities has increased, which has highlighted the health disparities that parallel this demographic. These disparities are reflected in the experience of transgender patients and their providers when seeking clinical laboratory services. CONTENT Little is known about the effect of gender-affirming hormone therapy and surgery on optimal laboratory test interpretation. Efforts to diminish health disparities encountered by transgender individuals and their providers can be accomplished by increasing social and clinical awareness regarding sex/gender incongruence and gaining insight into the physiological manifestations and laboratory interpretations of gender-affirming strategies. This review summarizes knowledge required to understand transgender healthcare including current clinical interventions for gender dysphoria. Particular attention is paid to the subsequent impact of these interventions on laboratory test utilization and interpretation. Common nomenclature and system barriers are also discussed. SUMMARY Understanding gender incongruence, the clinical changes associated with gender transition, and systemic barriers that maintain a gender/sex binary are key to providing adequate healthcare to transgender community. Transgender appropriate reference interval studies are virtually absent within the medical literature and should be explored. The laboratory has an important role in improving the physiological understanding, electronic medical system recognition, and overall social awareness of the transgender community.
Collapse
Affiliation(s)
- Zil Goldstein
- Department of Medical Education, Icahn School of Medicine, New York, NY
| | - Trevor A Corneil
- School of Population and Public Health, University of British Columbia, Vancouver, BC, Canada;
| | - Dina N Greene
- Department of Laboratory Medicine. University of Washington, Seattle, WA.
| |
Collapse
|
65
|
Maslah N, Cassinat B, Verger E, Kiladjian JJ, Velazquez L. The role of LNK/SH2B3 genetic alterations in myeloproliferative neoplasms and other hematological disorders. Leukemia 2017; 31:1661-1670. [PMID: 28484264 DOI: 10.1038/leu.2017.139] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 04/10/2017] [Accepted: 04/24/2017] [Indexed: 12/11/2022]
Abstract
Malignant hematological diseases are mainly because of the occurrence of molecular abnormalities leading to the deregulation of signaling pathways essential for precise cell behavior. High-resolution genome analysis using microarray and large-scale sequencing have helped identify several important acquired gene mutations that are responsible for such signaling deregulations across different hematological malignancies. In particular, the genetic landscape of classical myeloproliferative neoplasms (MPNs) has been in large part completed with the identification of driver mutations (targeting the cytokine receptor/Janus-activated kinase 2 (JAK2) pathway) that determine MPN phenotype, as well as additional mutations mainly affecting the regulation of gene expression (epigenetics or splicing regulators) and signaling. At present, most efforts concentrate in understanding how all these genetic alterations intertwine together to influence disease evolution and/or dictate clinical phenotype in order to use them to personalize diagnostic and clinical care. However, it is now evident that factors other than somatic mutations also play an important role in MPN disease initiation and progression, among which germline predisposition (single-nucleotide polymorphisms and haplotypes) may strongly influence the occurrence of MPNs. In this context, the LNK inhibitory adaptor protein encoded by the LNK/SH2B adaptor protein 3 (SH2B3) gene is the target of several genetic variations, acquired or inherited in MPNs, lymphoid leukemia and nonmalignant hematological diseases, underlying its importance in these pathological processes. As LNK adaptor is a key regulator of normal hematopoiesis, understanding the consequences of LNK variants on its protein functions and on driver or other mutations could be helpful to correlate genotype and phenotype of patients and to develop therapeutic strategies to target this molecule. In this review we summarize the current knowledge of LNK function in normal hematopoiesis, the different SH2B3 mutations reported to date and discuss how these genetic variations may influence the development of hematological malignancies.
Collapse
Affiliation(s)
- N Maslah
- APHP, Laboratoire de Biologie Cellulaire, Hôpital Saint-Louis, Paris, France.,Inserm UMRS 1131, IUH, Université Paris-Diderot, Paris, France
| | - B Cassinat
- APHP, Laboratoire de Biologie Cellulaire, Hôpital Saint-Louis, Paris, France.,Inserm UMRS 1131, IUH, Université Paris-Diderot, Paris, France
| | - E Verger
- APHP, Laboratoire de Biologie Cellulaire, Hôpital Saint-Louis, Paris, France.,Inserm UMRS 1131, IUH, Université Paris-Diderot, Paris, France
| | - J-J Kiladjian
- Inserm UMRS 1131, IUH, Université Paris-Diderot, Paris, France.,APHP, Centre d'investigations Cliniques, Hôpital Saint-Louis, Paris, France
| | - L Velazquez
- INSERM UMRS-MD1197, Institut André Lwoff/Université Paris XI, Hôpital Paul Brousse, Villejuif, France
| |
Collapse
|
66
|
Beyond immune thrombocytopenia: the evolving role of thrombopoietin receptor agonists. Ann Hematol 2017; 96:1421-1434. [PMID: 28275823 DOI: 10.1007/s00277-017-2953-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 02/13/2017] [Indexed: 01/19/2023]
Abstract
Since its discovery, the thrombopoietin (TPO) pathway has been an important pharmaceutical target for the treatment of thrombocytopenia. The first generation of TPO mimetics included peptide agents sharing homology with endogenous TPO, but these introduced a risk of antibody formation to endogenous TPO and were not successful. However, second-generation TPO mimetics or TPO receptor agonists (RAs) are currently being used to treat thrombocytopenia associated with a number of conditions, such as immune thrombocytopenia (ITP), severe aplastic anaemia (SAA), and hepatitis C virus-associated chronic liver disease. Accumulating efficacy and safety data suggest that the role of TPO-RAs in the treatment of thrombocytopenia may evolve in the near future with broader use of these agents in ITP and SAA, as well as approval in other indications, potentially including myelodysplastic syndromes, chemotherapy-associated thrombocytopenia, and post-transplant thrombocytopenia. This review provides an overview of clinical data on the efficacy and safety of TPO-RAs, emphasising recent findings that may expand their clinical utility.
Collapse
|
67
|
Zamora C, Cantó E, Nieto JC, Bardina J, Diaz-Torné C, Moya P, Magallares B, Ortiz MA, Julià G, Juarez C, Llobet JM, Vidal S. Binding of Platelets to Lymphocytes: A Potential Anti-Inflammatory Therapy in Rheumatoid Arthritis. THE JOURNAL OF IMMUNOLOGY 2017; 198:3099-3108. [PMID: 28250158 DOI: 10.4049/jimmunol.1601708] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 02/03/2017] [Indexed: 12/15/2022]
Abstract
Soluble factors released from platelets can modulate the immune response of leukocytes. We and others have recently found that T lymphocytes with bound platelets have reduced proliferation and IFN-γ and IL-17 production. Thus, we speculate that if we induce the binding of platelets to lymphocytes, we will be able to regulate the inflammatory response. When we cocultured platelets with lymphocytes at different ratios, we were able to increase the percentage of lymphocytes with bound platelets. The coculture of platelets with lymphocytes in the presence of stimulation decreased the production of IFN-γ and TNF-α, T cell proliferation, and the expression of CD25, PD-L1, and SLAM. However, this coculture increased CD39 expression. All of these effects were dependent on the dose of platelets and operated indistinctly with platelets from different healthy donors. When platelets were cocultured in the same compartment with lymphocytes, we observed less IFN-γ and TNF-α production and T lymphocyte proliferation than in cultures with platelets separated from lymphocytes by a 0.4-μm pore size filter. The binding of platelets to lymphocytes was blocked with anti-P-selectin Abs, and when this occurred we observed higher IFN-γ and TNF-α production than in nonblocked conditions. The cocultures of platelets with synovial fluid cells from rheumatoid arthritis patients reduced inflammatory cytokine production and increased IL-10 production. These results suggest that platelet binding to lymphocytes effectively regulates T lymphocyte function. This mechanism could be easily applied to reduce inflammatory responses.
Collapse
Affiliation(s)
- Carlos Zamora
- Departament Immunologia, Institut Recerca Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain;
| | - Elisabet Cantó
- Departament Immunologia, Institut Recerca Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Juan C Nieto
- Departament Immunologia, Institut Recerca Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Jorge Bardina
- Departament Immunologia, Institut Recerca Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Cesar Diaz-Torné
- Departament Immunologia, Institut Recerca Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain.,Unitat Reumatologia, Hospital de la Santa Creu i Sant Pau, 08026 Barcelona, Spain; and
| | - Patricia Moya
- Unitat Reumatologia, Hospital de la Santa Creu i Sant Pau, 08026 Barcelona, Spain; and
| | - Berta Magallares
- Unitat Reumatologia, Hospital de la Santa Creu i Sant Pau, 08026 Barcelona, Spain; and
| | - M Angels Ortiz
- Departament Immunologia, Institut Recerca Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Germà Julià
- Departament Immunologia, Institut Recerca Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - Candido Juarez
- Departament Immunologia, Hospital de la Santa Creu i Sant Pau, 08026 Barcelona, Spain
| | - Josep M Llobet
- Unitat Reumatologia, Hospital de la Santa Creu i Sant Pau, 08026 Barcelona, Spain; and
| | - Silvia Vidal
- Departament Immunologia, Institut Recerca Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain;
| |
Collapse
|
68
|
Grainger JD, Thind S. A practical guide to the use of eltrombopag in children with chronic immune thrombocytopenia. Pediatr Hematol Oncol 2017; 34:73-89. [PMID: 28537785 DOI: 10.1080/08880018.2017.1313918] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
INTRODUCTION Pediatric immune thrombocytopenia (ITP) may be associated with significant burden on children and their parents/caregivers. Thrombopoietin (TPO) receptor agonists (RAs) have been used to treat adult patients with chronic ITP (cITP) for nearly a decade and following pediatric studies Eltrombopag has been recently approved for pediatric cITP in the United States and Europe. TPO-RA s may help reduce the risk of bleeding and the need for conventional ITP therapies. REVIEW In this review, the clinical data demonstrating the efficacy and safety of TPO-RAs in pediatric ITP are evaluated, key recommendations regarding safe administration of eltrombopag are provided, and potential future directions in management of pediatric ITP are discussed.
Collapse
Affiliation(s)
- John D Grainger
- a Royal Manchester Children's Hospital , Manchester , United Kingdom
| | - Sharon Thind
- a Royal Manchester Children's Hospital , Manchester , United Kingdom
| |
Collapse
|
69
|
Ansari J, Yun JW, Kompelli AR, Moufarrej YE, Alexander JS, Herrera GA, Shackelford RE. The liquid biopsy in lung cancer. Genes Cancer 2017; 7:355-367. [PMID: 28191282 PMCID: PMC5302037 DOI: 10.18632/genesandcancer.127] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The incidence of lung cancer has significantly increased over the last century, largely due to smoking, and remains the most common cause of cancer deaths worldwide. This is often due to lung cancer first presenting at late stages and a lack of curative therapeutic options at these later stages. Delayed diagnoses, inadequate tumor sampling, and lung cancer misdiagnoses are also not uncommon due to the limitations of the tissue biopsy. Our better understanding of the tumor microenvironment and the systemic actions of tumors, combined with the recent advent of the liquid biopsy, may allow molecular diagnostics to be done on circulating tumor markers, particularly circulating tumor DNA. Multiple liquid biopsy molecular methods are presently being examined to determine their efficacy as surrogates to the tumor tissue biopsy. This review will focus on new liquid biopsy technologies and how they may assist in lung cancer detection, diagnosis, and treatment.
Collapse
Affiliation(s)
- Junaid Ansari
- Feist Weiller Cancer Center, LSU Health Shreveport, LA, USA; Department of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport, LA, USA
| | - Jungmi W Yun
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport, LA, USA
| | | | | | - Jonathan S Alexander
- Department of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport, LA, USA
| | | | | |
Collapse
|
70
|
Li S, Cao W, Sun X. Role of Platelet Parameters on Neovascular Glaucoma: A Retrospective Case-Control Study in China. PLoS One 2016; 11:e0166893. [PMID: 27907042 PMCID: PMC5131990 DOI: 10.1371/journal.pone.0166893] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 11/04/2016] [Indexed: 01/21/2023] Open
Abstract
Purpose Retinal vein occlusion (RVO) and diabetic retinopathy (DR) are two major sight-threatening diseases which may lead to neovascular glaucoma (NVG). The aim of this study was to explore the association between platelet parameters and NVG. Methods A total of 185 subjects were enrolled for the study from January, 2012 to December, 2015 at the Eye-ENT Hospital of Fudan University. Patients include those with NVG secondary to RVO (RVO group, n = 38), patients with NVG secondary to DR (DR group, n = 47), diabetics mellitus without retinopathy (DM group, n = 52), and healthy individuals (control group, n = 48). A complete ophthalmological examination including visual field examination, A-scan ultrasound, Fundus photography, and measurement of platelet parameters were performed for NVG subjects. Results There was no statistical difference in the mean age and gender among the RVO, DR, and control groups (p>0.05). The mean level of platelet distribution width (PDW) was higher (p<0.001) in the RVO group (15.16±2.13fl) and DR group (16.17±1.66fl) when compared with the control group (13.77±2.99fl). The mean plateletcrit (PCT) value of the RVO group (0.229±0.063%) was also higher (p = 0.049) than the control group (0.199±0.045). In the DR group, mean platelet volume (MPV) value (10.72±1.57fl) was significantly higher (p = 0.002) than the control group (9.75±0.89fl). A similar trend was observed when platelet parameters were compared among the 3 groups with respect to age. The mean level of PDW was significantly higher (p<0.001) in the DR group (16.17±1.66fl) compared with the DM group (13.80±3.32fl). Stepwise multiple logistic regression analysis revealed that PDW (OR = 1.44, 95%CI = 1.149–1.805, p = 0.002) and MPV (OR = 1.503, 95%CI = 1.031–2.192, p = 0.034) were associated with the DR group, PDW (OR = 1.207, 95%CI = 1.010–1.443, p = 0.039) and PCT (OR = 1.663, 95%CI = 1.870–2.654, p = 0.036) were associated with the RVO group. Conclusion Our results suggest that increased PDW and MPV are associated with the NVG secondary to DR group, elevated PDW and PCT are associated with the RVO group. It indicates that platelets might be an important factor in the onset and/or development of NVG.
Collapse
Affiliation(s)
- Shengjie Li
- Department of Clinical Laboratory, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China
| | - Wenjun Cao
- Department of Clinical Laboratory, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China
- * E-mail: (WC); (XS)
| | - Xinghuai Sun
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
- Key Laboratory of Myopia, Ministry of Health (Fudan University), Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai, China
- * E-mail: (WC); (XS)
| |
Collapse
|
71
|
Abstract
The primary function of platelets is to patrol the vasculature and seal vessel breaches to limit blood loss. However, it is becoming increasingly clear that they also contribute to pathophysiological conditions like thrombosis, atherosclerosis, stroke and infection. Severe sepsis is a devastating disease that claims hundreds of thousands of lives every year in North America and is a major burden to the public health system. Platelet surface receptors like GPIb, αIIbβ3, TLR2 and TLR4 are involved in direct platelet-bacteria interactions. Plasma proteins like fibrinogen and vWF enable indirect interactions. Furthermore, platelet granules contain a plethora of proteins that modulate the immune response as well as microbicidal agents which can directly lyse bacteria. Bacterial toxins are potent platelet activators and can cause intravascular platelet aggregation. Platelets contribute to the antibacterial response of the host involving Kupffer cells, neutrophils and the complement system. In this review we summarize the current knowledge about platelet-bacteria interactions and highlight recent advances in the field.
Collapse
Affiliation(s)
- Carsten Deppermann
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Paul Kubes
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
72
|
Zeller Meidell K, Robinson R, Vieira-de-Abreu A, Gormley AJ, Ghandehari H, W Grainger D, A Campbell R. RGDfK-functionalized gold nanorods bind only to activated platelets. J Biomed Mater Res A 2016; 105:209-217. [PMID: 27648522 DOI: 10.1002/jbm.a.35902] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 08/15/2016] [Accepted: 09/14/2016] [Indexed: 12/12/2022]
Abstract
Integrin-targeting peptide RGDfK-labeled gold nanorods (GNR) seek to improve hyperthermia targeted to solid tumors by exploiting the known up-regulation of integrin αvβ3 cell membrane proteins on solid tumor vasculature surfaces. Tumor binding specificity might be expected since surrounding tissues and endothelial cells have limited numbers of these receptors. However, RGD peptide binding to many proteins is promiscuous, with known affinity to several families of cell integrin receptors, and also possible binding to platelets after intravenous infusion via a different integrin receptor, αIIbβ3, on platelets. Binding of RGDfK-targeted GNR could considerably impact platelet function, ultimately leading to increased risk of bleeding or thrombosis depending on the degree of interaction. We sought to determine if RGDfK-labeled GNR could interact with platelets and alter platelet function. Targeted and untargeted nanorods exhibited little interaction with resting platelets in platelet rich plasma (PRP) preparations. However, upon platelet activation, peptide-targeted nanorods bound actively to platelets. Addition of RGDfK-GNR to unactivated platelets had little effect on markers of platelet activation, indicating that RGDfK-nanorods were incapable of inducing platelet activation. We next tested whether activated platelet function was altered in the presence of peptide-targeted nanorods. Platelet aggregation in whole blood and PRP in the presence of targeted nanorods had no significant effect on platelet aggregation. These data suggest that RGDfK-GNR alone have little impact on platelet function in plasma. However, nonspecific nanorod binding may occur in vascular beds where activated platelets are normally cleared, such as the spleen and liver, producing a possible toxicity risk for these nanomaterials. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 209-217, 2017.
Collapse
Affiliation(s)
- Krystin Zeller Meidell
- Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, Salt Lake City, Utah, 84112
| | - Ryan Robinson
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, 84112.,Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, Utah, 84112
| | - Adriana Vieira-de-Abreu
- Program in Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, Utah, 84112
| | - Adam J Gormley
- Department of Bioengineering, University of Utah, Salt Lake City, Utah, 84112.,Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, Utah, 84112
| | - Hamidreza Ghandehari
- Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, Salt Lake City, Utah, 84112.,Department of Bioengineering, University of Utah, Salt Lake City, Utah, 84112.,Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, Utah, 84112
| | - David W Grainger
- Department of Pharmaceutics and Pharmaceutical Chemistry, Health Sciences, University of Utah, Salt Lake City, Utah, 84112.,Department of Bioengineering, University of Utah, Salt Lake City, Utah, 84112.,Center for Nanomedicine, Nano Institute of Utah, University of Utah, Salt Lake City, Utah, 84112
| | - Robert A Campbell
- Program in Molecular Medicine, School of Medicine, University of Utah, Salt Lake City, Utah, 84112
| |
Collapse
|
73
|
Eicher JD, Chami N, Kacprowski T, Nomura A, Chen MH, Yanek LR, Tajuddin SM, Schick UM, Slater AJ, Pankratz N, Polfus L, Schurmann C, Giri A, Brody JA, Lange LA, Manichaikul A, Hill WD, Pazoki R, Elliot P, Evangelou E, Tzoulaki I, Gao H, Vergnaud AC, Mathias RA, Becker DM, Becker LC, Burt A, Crosslin DR, Lyytikäinen LP, Nikus K, Hernesniemi J, Kähönen M, Raitoharju E, Mononen N, Raitakari OT, Lehtimäki T, Cushman M, Zakai NA, Nickerson DA, Raffield LM, Quarells R, Willer CJ, Peloso GM, Abecasis GR, Liu DJ, Deloukas P, Samani NJ, Schunkert H, Erdmann J, Fornage M, Richard M, Tardif JC, Rioux JD, Dube MP, de Denus S, Lu Y, Bottinger EP, Loos RJF, Smith AV, Harris TB, Launer LJ, Gudnason V, Velez Edwards DR, Torstenson ES, Liu Y, Tracy RP, Rotter JI, Rich SS, Highland HM, Boerwinkle E, Li J, Lange E, Wilson JG, Mihailov E, Mägi R, Hirschhorn J, Metspalu A, Esko T, Vacchi-Suzzi C, Nalls MA, Zonderman AB, Evans MK, Engström G, Orho-Melander M, Melander O, O'Donoghue ML, Waterworth DM, Wallentin L, White HD, Floyd JS, Bartz TM, Rice KM, Psaty BM, Starr JM, Liewald DCM, Hayward C, Deary IJ, Greinacher A, Völker U, Thiele T, Völzke H, van Rooij FJA, Uitterlinden AG, Franco OH, Dehghan A, Edwards TL, Ganesh SK, Kathiresan S, Faraday N, Auer PL, Reiner AP, Lettre G, Johnson AD. Platelet-Related Variants Identified by Exomechip Meta-analysis in 157,293 Individuals. Am J Hum Genet 2016; 99:40-55. [PMID: 27346686 PMCID: PMC5005441 DOI: 10.1016/j.ajhg.2016.05.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/03/2016] [Indexed: 12/13/2022] Open
Abstract
Platelet production, maintenance, and clearance are tightly controlled processes indicative of platelets' important roles in hemostasis and thrombosis. Platelets are common targets for primary and secondary prevention of several conditions. They are monitored clinically by complete blood counts, specifically with measurements of platelet count (PLT) and mean platelet volume (MPV). Identifying genetic effects on PLT and MPV can provide mechanistic insights into platelet biology and their role in disease. Therefore, we formed the Blood Cell Consortium (BCX) to perform a large-scale meta-analysis of Exomechip association results for PLT and MPV in 157,293 and 57,617 individuals, respectively. Using the low-frequency/rare coding variant-enriched Exomechip genotyping array, we sought to identify genetic variants associated with PLT and MPV. In addition to confirming 47 known PLT and 20 known MPV associations, we identified 32 PLT and 18 MPV associations not previously observed in the literature across the allele frequency spectrum, including rare large effect (FCER1A), low-frequency (IQGAP2, MAP1A, LY75), and common (ZMIZ2, SMG6, PEAR1, ARFGAP3/PACSIN2) variants. Several variants associated with PLT/MPV (PEAR1, MRVI1, PTGES3) were also associated with platelet reactivity. In concurrent BCX analyses, there was overlap of platelet-associated variants with red (MAP1A, TMPRSS6, ZMIZ2) and white (PEAR1, ZMIZ2, LY75) blood cell traits, suggesting common regulatory pathways with shared genetic architecture among these hematopoietic lineages. Our large-scale Exomechip analyses identified previously undocumented associations with platelet traits and further indicate that several complex quantitative hematological, lipid, and cardiovascular traits share genetic factors.
Collapse
Affiliation(s)
- John D Eicher
- Population Sciences Branch, National Heart Lung and Blood Institute, The Framingham Heart Study, Framingham, MA 01702, USA
| | - Nathalie Chami
- Department of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada; Montreal Heart Institute, Montréal, QC H1T 1C8, Canada
| | - Tim Kacprowski
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald and Ernst-Mortiz-Arndt University Greifswald, Greifswald 17475, Germany; DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Akihiro Nomura
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Division of Cardiovascular Medicine, Kanazawa University Graduate School of Medical Science, Kanazawa, Ishikawa 9200942, Japan
| | - Ming-Huei Chen
- Population Sciences Branch, National Heart Lung and Blood Institute, The Framingham Heart Study, Framingham, MA 01702, USA
| | - Lisa R Yanek
- Department of Medicine, Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Salman M Tajuddin
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Ursula M Schick
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andrew J Slater
- Genetics, Target Sciences, GlaxoSmithKline, Research Triangle Park, NC 27709, USA; OmicSoft Corporation, Cary, NC 27513, USA
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55454, USA
| | - Linda Polfus
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Claudia Schurmann
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; The Genetics of Obesity and Related Metabolic Traits Program, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ayush Giri
- Division of Epidemiology, Institute for Medicine and Public Health, Vanderbilt University, Nashville, TN 37235, USA
| | - Jennifer A Brody
- Department of Medicine, University of Washington, Seattle, WA 98101, USA
| | - Leslie A Lange
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Ani Manichaikul
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, USA
| | - W David Hill
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9JZ, UK; Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Raha Pazoki
- Department of Epidemiology, Erasmus MC, Rotterdam 3000, the Netherlands
| | - Paul Elliot
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London W2 1PG, UK
| | - Evangelos Evangelou
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London W2 1PG, UK; Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina 45110, Greece
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London W2 1PG, UK; Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina 45110, Greece
| | - He Gao
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London W2 1PG, UK
| | - Anne-Claire Vergnaud
- Department of Epidemiology and Biostatistics, MRC-PHE Centre for Environment and Health, School of Public Health, Imperial College London, London W2 1PG, UK
| | - Rasika A Mathias
- Department of Medicine, Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Divisions of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Diane M Becker
- Department of Medicine, Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Lewis C Becker
- Department of Medicine, Division of General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Divisions of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Amber Burt
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - David R Crosslin
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, WA 98105, USA
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33520, Finland; Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere 33514, Finland
| | - Kjell Nikus
- Department of Cardiology, Heart Center, Tampere University Hospital, Tampere 33521, Finland; University of Tampere, School of Medicine, Tampere 33514, Finland
| | - Jussi Hernesniemi
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33520, Finland; Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere 33514, Finland; Department of Cardiology, Heart Center, Tampere University Hospital, Tampere 33521, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University Hospital, Tampere 33521, Finland; Department of Clinical Physiology, University of Tampere, Tampere 33514, Finland
| | - Emma Raitoharju
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33520, Finland; Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere 33514, Finland
| | - Nina Mononen
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33520, Finland; Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere 33514, Finland
| | - Olli T Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku 20521, Finland; Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku 20520, Finland
| | - Terho Lehtimäki
- Department of Clinical Chemistry, Fimlab Laboratories, Tampere 33520, Finland; Department of Clinical Chemistry, University of Tampere School of Medicine, Tampere 33514, Finland
| | - Mary Cushman
- Departments of Medicine and Pathology, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | - Neil A Zakai
- Departments of Medicine and Pathology, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | - Deborah A Nickerson
- Department of Genome Sciences, University of Washington, Seattle, WA 98105, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Rakale Quarells
- Morehouse School of Medicine, Social Epidemiology Research Center, Cardiovascular Research Institute, Atlanta, GA 30310, USA
| | - Cristen J Willer
- Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI 48108, USA; Department of Computational Medicine and Bioinformatics, Department of Human Genetics, University of Michigan, Ann Arbor, MI 48108, USA; Department of Biostatistics, University of Michigan, Ann Arbor, MI 48108, USA
| | - Gina M Peloso
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA; Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Goncalo R Abecasis
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, MI 48108, USA
| | - Dajiang J Liu
- Department of Public Health Sciences, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | - Panos Deloukas
- William Harvey Research Institute, Queen Mary University London, London E1 4NS, UK; Princess Al-Jawhara Al-Brahim Centre of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester LE1 7RH, UK; NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, Leicester LE3 9QP, UK
| | - Heribert Schunkert
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich 80333, Germany; Deutsches Herzzentrum München, Technische Universität München, Munich 80333, Germany
| | - Jeanette Erdmann
- Institute for Integrative and Experimental Genomics, University of Lübeck, Lübeck 23562, Germany; DZHK (German Research Centre for Cardiovascular Research), partner site Hamburg/Lübeck/Kiel, Lübeck 23562, Germany
| | - Myriam Fornage
- Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Melissa Richard
- Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jean-Claude Tardif
- Department of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada; Montreal Heart Institute, Montréal, QC H1T 1C8, Canada
| | - John D Rioux
- Department of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada; Montreal Heart Institute, Montréal, QC H1T 1C8, Canada
| | - Marie-Pierre Dube
- Department of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada; Montreal Heart Institute, Montréal, QC H1T 1C8, Canada
| | - Simon de Denus
- Montreal Heart Institute, Montréal, QC H1T 1C8, Canada; Faculty of Pharmacy, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Yingchang Lu
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Erwin P Bottinger
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Albert Vernon Smith
- Icelandic Heart Association, Kopavogur 201, Iceland; Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Tamara B Harris
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Intramural Research Program, NIH, Bethesda, MD 21224, USA
| | - Lenore J Launer
- Laboratory of Epidemiology, Demography, and Biometry, National Institute on Aging, Intramural Research Program, NIH, Bethesda, MD 21224, USA
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur 201, Iceland; Faculty of Medicine, University of Iceland, Reykjavik 101, Iceland
| | - Digna R Velez Edwards
- Vanderbilt Epidemiology Center, Department of Obstetrics & Gynecology, Institute for Medicine and Public Health, Vanderbilt Genetics Institute, Vanderbilt University, Nashville, TN 37203, USA
| | - Eric S Torstenson
- Division of Epidemiology, Institute for Medicine and Public Health, Vanderbilt University, Nashville, TN 37235, USA
| | - Yongmei Liu
- Center for Human Genetics, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Russell P Tracy
- Departments of Pathology and Laboratory Medicine and Biochemistry, University of Vermont College of Medicine, Colchester, VT 05446, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles Biomedical Research Institute, Torrance, CA 90502, USA; Department of Pediatrics, Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, USA
| | - Heather M Highland
- The University of Texas School of Public Health, The University of Texas Graduate School of Biomedical Sciences at Houston, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Department of Epidemiology, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX 77030, USA; Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jin Li
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Ethan Lange
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27514, USA; Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Evelin Mihailov
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Reedik Mägi
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Joel Hirschhorn
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA; Department of Endocrinology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Andres Metspalu
- Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Tõnu Esko
- Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA; Estonian Genome Center, University of Tartu, Tartu 51010, Estonia
| | - Caterina Vacchi-Suzzi
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Mike A Nalls
- Laboratory of Neurogenetics, National Institute on Aging, NIH, Bethesda, MD 21224, USA
| | - Alan B Zonderman
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Michele K Evans
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, NIH, Baltimore, MD 21224, USA
| | - Gunnar Engström
- Department of Clinical Sciences Malmö, Lund University, Malmö 221 00, Sweden; Skåne University Hospital, Malmö 222 41, Sweden
| | - Marju Orho-Melander
- Department of Clinical Sciences Malmö, Lund University, Malmö 221 00, Sweden; Skåne University Hospital, Malmö 222 41, Sweden
| | - Olle Melander
- Department of Clinical Sciences Malmö, Lund University, Malmö 221 00, Sweden; Skåne University Hospital, Malmö 222 41, Sweden
| | - Michelle L O'Donoghue
- TIMI Study Group, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Dawn M Waterworth
- Genetics, Target Sciences, GlaxoSmithKline, King of Prussia, PA 19406, USA
| | - Lars Wallentin
- Department of Medical Sciences, Cardiology, and Uppsala Clinical Research Center, Uppsala University, Uppsala 751 85, Sweden
| | - Harvey D White
- Green Lane Cardiovascular Service, Auckland City Hospital and University of Auckland, Auckland 1142, New Zealand
| | - James S Floyd
- Department of Medicine, University of Washington, Seattle, WA 98101, USA
| | - Traci M Bartz
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Kenneth M Rice
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Departments of Medicine, Epidemiology and Health Services, University of Washington, Seattle, WA 98101, USA; Group Health Research Institute, Group Health Cooperative, Seattle, WA 98101, USA
| | - J M Starr
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9JZ, UK; Alzheimer Scotland Research Centre, Edinburgh EH8 9JZ, UK
| | - David C M Liewald
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9JZ, UK; Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh EH8 9JZ, UK; Department of Psychology, University of Edinburgh, Edinburgh EH8 9JZ, UK
| | - Andreas Greinacher
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald 17475, Germany
| | - Uwe Völker
- Department of Functional Genomics, Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald and Ernst-Mortiz-Arndt University Greifswald, Greifswald 17475, Germany; DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany
| | - Thomas Thiele
- Institute for Immunology and Transfusion Medicine, University Medicine Greifswald, Greifswald 17475, Germany
| | - Henry Völzke
- DZHK (German Centre for Cardiovascular Research), partner site Greifswald, Greifswald, Germany; Institute for Community Medicine, University Medicine Greifswald, Greifswald 13347, Germany
| | | | - André G Uitterlinden
- Department of Epidemiology, Erasmus MC, Rotterdam 3000, the Netherlands; Department of Internal Medicine, Erasmus MC, Rotterdam 3000, the Netherlands; Netherlands Consortium for Healthy Ageing (NCHA), Rotterdam 3015, the Netherlands
| | - Oscar H Franco
- Department of Epidemiology, Erasmus MC, Rotterdam 3000, the Netherlands
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus MC, Rotterdam 3000, the Netherlands
| | - Todd L Edwards
- Division of Epidemiology, Institute for Medicine and Public Health, Vanderbilt University, Nashville, TN 37235, USA
| | - Santhi K Ganesh
- Departments of Internal and Human Genetics, University of Michigan, Ann Arbor, MI 48108, USA
| | - Sekar Kathiresan
- Center for Human Genetic Research, Massachusetts General Hospital, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute, Cambridge, MA 02142, USA; Cardiovascular Research Center, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Nauder Faraday
- Department of Anesthesiology & Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Paul L Auer
- Zilber School of Public Health, University of Wisconsin-Milwaukee, Milwaukee, WI 53205, USA
| | - Alex P Reiner
- Department of Epidemiology, University of Washington, Seattle, WA 98105, USA; Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Guillaume Lettre
- Department of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada; Montreal Heart Institute, Montréal, QC H1T 1C8, Canada
| | - Andrew D Johnson
- Population Sciences Branch, National Heart Lung and Blood Institute, The Framingham Heart Study, Framingham, MA 01702, USA.
| |
Collapse
|
74
|
Li J, Sharkey CC, Wun B, Liesveld JL, King MR. Genetic engineering of platelets to neutralize circulating tumor cells. J Control Release 2016; 228:38-47. [PMID: 26921521 PMCID: PMC4828270 DOI: 10.1016/j.jconrel.2016.02.036] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 02/10/2016] [Accepted: 02/23/2016] [Indexed: 12/12/2022]
Abstract
Mounting experimental evidence demonstrates that platelets support cancer metastasis. Within the circulatory system, platelets guard circulating tumor cells (CTCs) from immune elimination and promote their arrest at the endothelium, supporting CTC extravasation into secondary sites. Neutralization of CTCs in blood circulation can potentially attenuate metastases to distant organs. Therefore, extensive studies have explored the blockade of platelet-CTC interactions as an anti-metastatic strategy. Such an intervention approach, however, may cause bleeding disorders since the platelet-CTC interactions inherently rely on the blood coagulation cascade including platelet activation. On the other hand, platelets have been genetically engineered to correct inherited bleeding disorders in both animal models and human clinical trials. In this study, inspired by the physical association between platelets and CTCs, platelets were genetically modified to express surface-bound tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), a cytokine known to induce apoptosis specifically in tumor cells. The TRAIL-expressing platelets were demonstrated to kill cancer cells in vitro and significantly reduce metastases in a mouse model of prostate cancer metastasis. Our results suggest that using platelets to produce and deliver cancer-specific therapeutics can provide a Trojan-horse strategy of neutralizing CTCs to attenuate metastasis.
Collapse
Affiliation(s)
- Jiahe Li
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Charles C Sharkey
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Brittany Wun
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Jane L Liesveld
- Department of Medicine, Hematology/Oncology (SMD), University of Rochester Medical Center, School of Medicine and Dentistry, Rochester, NY 14642, USA
| | - Michael R King
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
75
|
Du J, Wang Q, He B, Liu P, Chen JY, Quan H, Ma X. Association of mean platelet volume and platelet count with the development and prognosis of ischemic and hemorrhagic stroke. Int J Lab Hematol 2016; 38:233-9. [PMID: 26992440 DOI: 10.1111/ijlh.12474] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 12/31/2015] [Indexed: 11/30/2022]
Affiliation(s)
- J. Du
- Department of Neurology; The Nuclear Industry 416 Hospital; Chengdu Sichuan China
- Department of Health and Social Behavior; West China School of Public Health, Sichuan University; Chengdu Sichuan China
| | - Q. Wang
- Department of Neurology; The Nuclear Industry 416 Hospital; Chengdu Sichuan China
| | - B. He
- Department of Health and Social Behavior; West China School of Public Health, Sichuan University; Chengdu Sichuan China
| | - P. Liu
- Department of Health and Social Behavior; West China School of Public Health, Sichuan University; Chengdu Sichuan China
| | - J.-Y. Chen
- Department of Health and Social Behavior; West China School of Public Health, Sichuan University; Chengdu Sichuan China
| | - H. Quan
- Department of Neurology; The Nuclear Industry 416 Hospital; Chengdu Sichuan China
| | - X. Ma
- Department of Neurology; The Nuclear Industry 416 Hospital; Chengdu Sichuan China
| |
Collapse
|
76
|
Vázquez-Santiago M, Ziyatdinov A, Pujol-Moix N, Brunel H, Morera A, Soria JM, Souto JC. Age and gender effects on 15 platelet phenotypes in a Spanish population. Comput Biol Med 2016; 69:226-33. [PMID: 26773944 DOI: 10.1016/j.compbiomed.2015.12.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/22/2015] [Accepted: 12/26/2015] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Several studies have analysed the platelet parameters in human blood, nevertheless there are no extensive analyses on the less common platelet phenotypes. The main objective of our study is to evaluate the age and gender effects on 15 platelet phenotypes. METHODS We studied 804 individuals, ranging in age from 2 to 93 years, included in the Genetic Analysis of Idiopathic Thrombophilia 2 (GAIT 2) Project. The 15 platelet phenotypes analysed were the platelets counts, platelet volumes, plateletcrits, immature platelet fraction (IPF) and platelet function assay (PFA). A regression-based method was used to evaluate the age and gender effects on these phenotypes. RESULTS Our results were consistent with the previously reported results regarding platelet counts and plateletcrit (PCT). They showed a decrease with increasing age. The mean platelet volume (MPV), platelet distribution width (PDW) and platelet-large cell ratio (P-LCR) increased with age, but did not present any gender effect. All the IPF phenotypes increased with age, whereas the PFA phenotypes did not show any relation to age or gender. DISCUSSION To sum up, our study provides a comprehensive analysis of the age and gender effects on the platelet phenotypes in a family-base sample. Our results suggest more reasonable age stratification into two distinct groups: childhood, ranging from 2 to 12 years, and the mature group, from 13 to 93 years. Moreover, the PFA phenotypes were maintained constant while the platelet counts, the MPV and IPF levels vary with age.
Collapse
Affiliation(s)
- Miquel Vázquez-Santiago
- Unitat d'Hemostàsia i Trombosi, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret, 167, Barcelona 08025, Spain.
| | - Andrey Ziyatdinov
- Unit of Genomics of Complex Diseases, Sant Pau Institute of Biomedical Research (IIB-Sant Pau), Sant Antoni Maria Claret, 167, Barcelona 08025, Spain.
| | - Núria Pujol-Moix
- Unitat d'Hemostàsia i Trombosi, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret, 167, Barcelona 08025, Spain; Department of Medicine, Universitat Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret, 167, Barcelona 08025, Spain.
| | - Helena Brunel
- Unit of Genomics of Complex Diseases, Sant Pau Institute of Biomedical Research (IIB-Sant Pau), Sant Antoni Maria Claret, 167, Barcelona 08025, Spain.
| | - Agnès Morera
- Unitat d'Hemostàsia i Trombosi, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret, 167, Barcelona 08025, Spain.
| | - José Manuel Soria
- Unit of Genomics of Complex Diseases, Sant Pau Institute of Biomedical Research (IIB-Sant Pau), Sant Antoni Maria Claret, 167, Barcelona 08025, Spain.
| | - Juan Carlos Souto
- Unitat d'Hemostàsia i Trombosi, Hospital de la Santa Creu i Sant Pau, Sant Antoni Maria Claret, 167, Barcelona 08025, Spain.
| |
Collapse
|
77
|
Breen MS, Maihofer AX, Glatt SJ, Tylee DS, Chandler SD, Tsuang MT, Risbrough VB, Baker DG, O’Connor DT, Nievergelt CM, Woelk CH. Gene networks specific for innate immunity define post-traumatic stress disorder. Mol Psychiatry 2015; 20:1538-45. [PMID: 25754082 PMCID: PMC4565790 DOI: 10.1038/mp.2015.9] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 11/25/2014] [Accepted: 12/19/2014] [Indexed: 12/17/2022]
Abstract
The molecular factors involved in the development of Post-Traumatic Stress Disorder (PTSD) remain poorly understood. Previous transcriptomic studies investigating the mechanisms of PTSD apply targeted approaches to identify individual genes under a cross-sectional framework lack a holistic view of the behaviours and properties of these genes at the system-level. Here we sought to apply an unsupervised gene-network based approach to a prospective experimental design using whole-transcriptome RNA-Seq gene expression from peripheral blood leukocytes of U.S. Marines (N=188), obtained both pre- and post-deployment to conflict zones. We identified discrete groups of co-regulated genes (i.e., co-expression modules) and tested them for association to PTSD. We identified one module at both pre- and post-deployment containing putative causal signatures for PTSD development displaying an over-expression of genes enriched for functions of innate-immune response and interferon signalling (Type-I and Type-II). Importantly, these results were replicated in a second non-overlapping independent dataset of U.S. Marines (N=96), further outlining the role of innate immune and interferon signalling genes within co-expression modules to explain at least part of the causal pathophysiology for PTSD development. A second module, consequential of trauma exposure, contained PTSD resiliency signatures and an over-expression of genes involved in hemostasis and wound responsiveness suggesting that chronic levels of stress impair proper wound healing during/after exposure to the battlefield while highlighting the role of the hemostatic system as a clinical indicator of chronic-based stress. These findings provide novel insights for early preventative measures and advanced PTSD detection, which may lead to interventions that delay or perhaps abrogate the development of PTSD.
Collapse
Affiliation(s)
- Michael S. Breen
- Clinical and Experimental Sciences, Faculty of Medicine,
University of Southampton, UK
| | - Adam X. Maihofer
- Department of Psychiatry, University of California San
Diego, California, USA
| | - Stephen J. Glatt
- Psychiatric Genetic Epidemiology and Neurobiology
Laboratory (PsychGENe Lab), Departments of Psychiatry and Behavioral Sciences and
Neuroscience and Physiology, Medical Genetics Research Center, SUNY Upstate Medical
University, Syracuse, New York, USA
| | - Daniel S. Tylee
- Psychiatric Genetic Epidemiology and Neurobiology
Laboratory (PsychGENe Lab), Departments of Psychiatry and Behavioral Sciences and
Neuroscience and Physiology, Medical Genetics Research Center, SUNY Upstate Medical
University, Syracuse, New York, USA
| | - Sharon D. Chandler
- Department of Psychiatry, University of California San
Diego, California, USA
| | - Ming T. Tsuang
- Department of Psychiatry, University of California San
Diego, California, USA,Veterans Affairs Center of Excellence for Stress and Mental
Health, San Diego, California, USA,Veterans Affairs San Diego Healthcare System, San Diego,
California, USA,Institute of Genomic Medicine, University of California,
San Diego, La Jolla, California USA,Center for Behavioral Genomics, Department of Psychiatry,
University of California San Diego, California, USA
| | - Victoria B. Risbrough
- Department of Psychiatry, University of California San
Diego, California, USA,Veterans Affairs Center of Excellence for Stress and Mental
Health, San Diego, California, USA
| | - Dewleen G. Baker
- Department of Psychiatry, University of California San
Diego, California, USA,Veterans Affairs Center of Excellence for Stress and Mental
Health, San Diego, California, USA
| | - Daniel T. O’Connor
- Institute of Genomic Medicine, University of California,
San Diego, La Jolla, California USA,Departments of Medicine and Pharmacology, University of
California San Diego, California, USA
| | - Caroline M. Nievergelt
- Department of Psychiatry, University of California San
Diego, California, USA,Veterans Affairs Center of Excellence for Stress and Mental
Health, San Diego, California, USA
| | - Christopher H. Woelk
- Clinical and Experimental Sciences, Faculty of Medicine,
University of Southampton, UK
| |
Collapse
|
78
|
Mauler M, Seyfert J, Haenel D, Seeba H, Guenther J, Stallmann D, Schoenichen C, Hilgendorf I, Bode C, Ahrens I, Duerschmied D. Platelet-neutrophil complex formation-a detailed in vitro analysis of murine and human blood samples. J Leukoc Biol 2015; 99:781-9. [PMID: 26578648 DOI: 10.1189/jlb.3ta0315-082r] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 10/28/2015] [Indexed: 12/13/2022] Open
Abstract
Platelets form complexes with neutrophils during inflammatory processes. These aggregates migrate into affected tissues and also circulate within the organism. Several studies have evaluated platelet-neutrophil complexes as a marker of cardiovascular diseases in human and mouse. Although multiple publications have reported platelet-neutrophil complex counts, we noticed that different methods were used to analyze platelet-neutrophil complex formation, resulting in significant differences, even in baseline values. We established a protocol for platelet-neutrophil complex measurement with flow cytometry in murine and human whole blood samples. In vitro platelet-neutrophil complex formation was stimulated with ADP or PMA. We tested the effect of different sample preparation steps and cytometer settings on platelet-neutrophil complex detection and noticed false-positive counts with increasing acquisition speed. Platelet-neutrophil complex formation depends on platelet P-selectin expression, and antibody blocking of P-selectin consequently prevented ADP-induced platelet-neutrophil complex formation. These findings may help generating more comparable data among different research groups that examine platelet-neutrophil complexes as a marker for cardiovascular disease and novel therapeutic interventions.
Collapse
Affiliation(s)
- Maximilian Mauler
- Faculty of Biology, Heart Center, University of Freiburg, Germany and Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Julia Seyfert
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - David Haenel
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Hannah Seeba
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Janine Guenther
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Daniela Stallmann
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Claudia Schoenichen
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Ingo Hilgendorf
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Christoph Bode
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Ingo Ahrens
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| | - Daniel Duerschmied
- Department of Cardiology and Angiology I, Heart Center, University of Freiburg, Germany
| |
Collapse
|
79
|
Adeli K, Raizman JE, Chen Y, Higgins V, Nieuwesteeg M, Abdelhaleem M, Wong SL, Blais D. Complex Biological Profile of Hematologic Markers across Pediatric, Adult, and Geriatric Ages: Establishment of Robust Pediatric and Adult Reference Intervals on the Basis of the Canadian Health Measures Survey. Clin Chem 2015; 61:1075-86. [DOI: 10.1373/clinchem.2015.240531] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 05/06/2015] [Indexed: 11/06/2022]
Abstract
AbstractBACKGROUNDIn a collaboration between the Canadian Laboratory Initiative on Pediatric Reference Intervals (CALIPER) and the Canadian Health Measures Survey (CHMS), we determined reference value distributions using an a priori approach and created a comprehensive database of age- and sex-stratified reference intervals for clinically relevant hematologic parameters in a large household population of children and adults.METHODSThe CHMS collected data and blood samples from 11 999 respondents aged 3–79 years. Hematology markers were measured with either the Beckman Coulter HmX or Siemens Sysmex CA-500 Series analyzers. After applying exclusion criteria and removing outliers, we determined statistically relevant age and sex partitions and calculated reference intervals, including 90% CIs, according to CSLI C28-A3 guidelines.RESULTSHematology marker values showed dynamic changes from childhood into adulthood as well as between sexes, necessitating distinct partitions throughout life. Most age partitions were necessary during childhood, reflecting the hematologic changes that occur during growth and development. Hemoglobin, red blood cell count, hematocrit, and indices (mean corpuscular volume, mean corpuscular hemoglobin, and mean corpuscular hemoglobin concentration) increased with age, but females had lower hemoglobin and hematocrit starting at puberty. Platelet count gradually decreased with age and required multiple sex partitions during adolescence and adulthood. White blood cell count remained relatively constant over life, whereas fibrinogen increased slightly, requiring distinct age and sex partitions.CONCLUSIONSThe robust dataset generated in this study has allowed observation of dynamic biological profiles of several hematology markers and the establishment of comprehensive age- and sex-specific reference intervals that may contribute to accurate monitoring of pediatric, adult, and geriatric patients.
Collapse
Affiliation(s)
- Khosrow Adeli
- CALIPER Program, Divisions of Clinical Biochemistry and Hematopathology, Pediatric Laboratory Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Joshua E Raizman
- CALIPER Program, Divisions of Clinical Biochemistry and Hematopathology, Pediatric Laboratory Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Yunqi Chen
- CALIPER Program, Divisions of Clinical Biochemistry and Hematopathology, Pediatric Laboratory Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Victoria Higgins
- CALIPER Program, Divisions of Clinical Biochemistry and Hematopathology, Pediatric Laboratory Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Michelle Nieuwesteeg
- CALIPER Program, Divisions of Clinical Biochemistry and Hematopathology, Pediatric Laboratory Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Mohamed Abdelhaleem
- CALIPER Program, Divisions of Clinical Biochemistry and Hematopathology, Pediatric Laboratory Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Suzy L Wong
- Health Analysis Division, Statistics Canada, Ottawa, ON
| | - David Blais
- Health Statistics Division, Statistics Canada, Ottawa, ON
- Current affiliation: Section Head, Laboratory Services, Health Canada, Ottawa, ON, Canada
| |
Collapse
|
80
|
Revisiting the case for genetically engineered mouse models in human myelodysplastic syndrome research. Blood 2015; 126:1057-68. [PMID: 26077396 DOI: 10.1182/blood-2015-01-624239] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 06/01/2015] [Indexed: 01/11/2023] Open
Abstract
Much-needed attention has been given of late to diseases specifically associated with an expanding elderly population. Myelodysplastic syndrome (MDS), a hematopoietic stem cell-based blood disease, is one of these. The lack of clear understanding of the molecular mechanisms underlying the pathogenesis of this disease has hampered the development of efficacious therapies, especially in the presence of comorbidities. Mouse models could potentially provide new insights into this disease, although primary human MDS cells grow poorly in xenografted mice. This makes genetically engineered murine models a more attractive proposition, although this approach is not without complications. In particular, it is unclear if or how myelodysplasia (abnormal blood cell morphology), a key MDS feature in humans, presents in murine cells. Here, we evaluate the histopathologic features of wild-type mice and 23 mouse models with verified myelodysplasia. We find that certain features indicative of myelodysplasia in humans, such as Howell-Jolly bodies and low neutrophilic granularity, are commonplace in healthy mice, whereas other features are similarly abnormal in humans and mice. Quantitative hematopoietic parameters, such as blood cell counts, are required to distinguish between MDS and related diseases. We provide data that mouse models of MDS can be genetically engineered and faithfully recapitulate human disease.
Collapse
|
81
|
Kulshrestha M, Sola-Visner M, Widness JA, Veng-Pedersen P, Mager DE. Mathematical model of platelet turnover in thrombocytopenic and nonthrombocytopenic preterm neonates. Am J Physiol Heart Circ Physiol 2014; 308:H68-73. [PMID: 25362135 DOI: 10.1152/ajpheart.00528.2013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Neonatal thrombocytopenia affects 22-35% of all neonates admitted to neonatal intensive care units. The purpose of this study was to develop a mathematical model for characterizing platelet (PLT) kinetics in thrombocytopenic preterm neonates. Immature PLT fraction (IPF) and PLT counts were measured for up to 35 days after birth in 27 very low birth weight preterm neonates. PLT transfusions were administered to 8 of the 27 (24%) subjects. The final model included a series of four transit compartments to mimic the production and survival of IPF and PLT. Model parameters were estimated using nonlinear mixed effects modeling with the maximum likelihood expectation maximization algorithm. The model adequately captured the diverse phenotypes expressed by individual subject profiles. Typical population survival values for IPF and PLT life spans in nonthrombocytopenic patients were estimated at 0.912 and 10.7 days, respectively. These values were significantly shorter in thrombocytopenic subjects, 0.429 and 2.56 days, respectively. The model was also used to evaluate the influence of growth and laboratory phlebotomy loss on the time course of IPF and PLT counts. Whereas incorporating body weight was essential to correct for expanding blood volume due to growth, phlebotomy loss, a possible covariate, did not significantly influence PLT kinetics. This study provides a platform for identifying potential covariates that influence the interindividual variability in model parameters regulating IPF and PLT kinetics and for evaluating future pharmacological therapies for treating thrombocytopenic neonates.
Collapse
Affiliation(s)
- Mudit Kulshrestha
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York
| | - Martha Sola-Visner
- Division of Newborn Medicine, Children's Hospital Boston, Boston, Massachusetts
| | - John A Widness
- Department of Pediatrics, College of Medicine, University of Iowa, Iowa City, Iowa; and
| | | | - Donald E Mager
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York;
| |
Collapse
|
82
|
Han X, Zhang L, Zhang Z, Zhang Z, Wang J, Yang J, Niu J. Association between phosphatase related gene variants and coronary artery disease: case-control study and meta-analysis. Int J Mol Sci 2014; 15:14058-76. [PMID: 25123136 PMCID: PMC4159839 DOI: 10.3390/ijms150814058] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 07/21/2014] [Accepted: 07/30/2014] [Indexed: 12/26/2022] Open
Abstract
Recent studies showed that the serum alkaline phosphatase is an independent predictor of the coronary artery disease (CAD). In this work, we aimed to summarize the association between three phosphatase related single nucleotide polymorphisms (rs12526453, rs11066301 and rs3828329) and the risk of CAD in Han Chinese. Our results showed that the rs3828329 of the ACP1 gene was closely related to the risk of CAD in Han Chinese (OR = 1.45, p = 0.0006). This significant association of rs3828329 with CAD was only found in the females (Additive model: OR = 1.80, p = 0.001; dominant model: OR = 1.69, p = 0.03; recessive model: OR = 1.96, p = 0.0008). Moreover, rs3828329 was likely to exert its effect in females aged 65 years and older (OR = 2.27, p = 0.001). Further meta-analyses showed that the rs12526453 of PHACTR11 gene (OR = 1.14, p < 0.0001, random-effect method) and the rs11066301 of PTPN11 gene (OR = 1.15, p < 0.0001, fixed-effects method) were associated with CAD risk in multiple populations. Our results showed that the polymorphisms rs12526453 and rs11066301 are significantly associated with the CAD risk in multiple populations. The rs3828329 of ACP1 gene is also a risk factor of CAD in Han Chinese females aged 65 years and older.
Collapse
Affiliation(s)
- Xia Han
- Department of Cardiology, Laiwu People's Hospital, Laiwu 271100, China.
| | - Lijun Zhang
- Department of Cardiology, Laiwu People's Hospital, Laiwu 271100, China.
| | - Zhiqiang Zhang
- Department of Cardiology, Laiwu People's Hospital, Laiwu 271100, China.
| | - Zengtang Zhang
- Department of Cardiology, Laiwu People's Hospital, Laiwu 271100, China.
| | - Jianchun Wang
- Department of Cardiology, Shandong Provincial Hospital, Jinan 250000, China.
| | - Jun Yang
- Department of Cardiology, Yantai Yuhuangding Hospital, Yantai 264000, China.
| | - Jiamin Niu
- Department of Cardiology, Laiwu People's Hospital, Laiwu 271100, China.
| |
Collapse
|
83
|
Shameer K, Denny JC, Ding K, Jouni H, Crosslin DR, de Andrade M, Chute CG, Peissig P, Pacheco JA, Li R, Bastarache L, Kho AN, Ritchie MD, Masys DR, Chisholm RL, Larson EB, McCarty CA, Roden DM, Jarvik GP, Kullo IJ. A genome- and phenome-wide association study to identify genetic variants influencing platelet count and volume and their pleiotropic effects. Hum Genet 2014; 133:95-109. [PMID: 24026423 PMCID: PMC3880605 DOI: 10.1007/s00439-013-1355-7] [Citation(s) in RCA: 108] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 08/22/2013] [Indexed: 12/21/2022]
Abstract
Platelets are enucleated cell fragments derived from megakaryocytes that play key roles in hemostasis and in the pathogenesis of atherothrombosis and cancer. Platelet traits are highly heritable and identification of genetic variants associated with platelet traits and assessing their pleiotropic effects may help to understand the role of underlying biological pathways. We conducted an electronic medical record (EMR)-based study to identify common variants that influence inter-individual variation in the number of circulating platelets (PLT) and mean platelet volume (MPV), by performing a genome-wide association study (GWAS). We characterized genetic variants associated with MPV and PLT using functional, pathway and disease enrichment analyses; we assessed pleiotropic effects of such variants by performing a phenome-wide association study (PheWAS) with a wide range of EMR-derived phenotypes. A total of 13,582 participants in the electronic MEdical Records and GEnomic network had data for PLT and 6,291 participants had data for MPV. We identified five chromosomal regions associated with PLT and eight associated with MPV at genome-wide significance (P < 5E-8). In addition, we replicated 20 SNPs [out of 56 SNPs (α: 0.05/56 = 9E-4)] influencing PLT and 22 SNPs [out of 29 SNPs (α: 0.05/29 = 2E-3)] influencing MPV in a published meta-analysis of GWAS of PLT and MPV. While our GWAS did not find any new associations, our functional analyses revealed that genes in these regions influence thrombopoiesis and encode kinases, membrane proteins, proteins involved in cellular trafficking, transcription factors, proteasome complex subunits, proteins of signal transduction pathways, proteins involved in megakaryocyte development, and platelet production and hemostasis. PheWAS using a single-SNP Bonferroni correction for 1,368 diagnoses (0.05/1368 = 3.6E-5) revealed that several variants in these genes have pleiotropic associations with myocardial infarction, autoimmune, and hematologic disorders. We conclude that multiple genetic loci influence interindividual variation in platelet traits and also have significant pleiotropic effects; the related genes are in multiple functional pathways including those relevant to thrombopoiesis.
Collapse
Affiliation(s)
- Khader Shameer
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | - Joshua C. Denny
- Departments of Medicine and Biomedical Informatics, Vanderbilt University, Nashville, TN 37232, USA
| | - Keyue Ding
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | - Hayan Jouni
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| | - David R. Crosslin
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Mariza de Andrade
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Christopher G. Chute
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Peggy Peissig
- Biomedical Informatics Research Center, Marshfield Clinic, Marshfield, WI, 54449, USA
| | - Jennifer A. Pacheco
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Rongling Li
- Office of Population Genomics, National Human Genome Research Institute, 5635 Fishers Lane, Suite 3058, MSC 9307, Bethesda, MD, 20892, USA
| | - Lisa Bastarache
- Departments of Medicine and Biomedical Informatics, Vanderbilt University, Nashville, TN 37232, USA
| | - Abel N. Kho
- Department of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Marylyn D Ritchie
- Center for Systems Genomics, Pennsylvania State University, Eberly College of Science, The Huck Institutes of the Life Sciences, 512 Wartik Laboratory, University Park, PA 16802 USA
| | - Daniel R. Masys
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Room 416 Eskind Medical Library, Nashville, TN, 37232, USA
| | - Rex L. Chisholm
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Eric B. Larson
- Group Health Research Institute, 1730 Minor Avenue, Suite 1600, Seattle, WA, 98101, USA
| | | | - Dan M. Roden
- Department of Pharmacology, Vanderbilt University School of Medicine, 1285 Medical Research Building IV, Nashville, TN, 37232, USA
| | - Gail P. Jarvik
- Department of Genome Sciences, University of Washington, 3720 15th Ave NE, Seattle WA 98195, USA
| | - Iftikhar J. Kullo
- Division of Cardiovascular Diseases, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
84
|
Gallerani M, Reverberi R, Salmi R, Smolensky MH, Manfredini R. Seasonal variation of platelets in a cohort of Italian blood donors: a preliminary report. Eur J Med Res 2013; 18:31. [PMID: 24044785 PMCID: PMC3848585 DOI: 10.1186/2047-783x-18-31] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 09/10/2013] [Indexed: 11/17/2022] Open
Abstract
Background Since available data are not univocal, the aim of this study was to explore the existence of a seasonal variation in platelet count. Methods The study was based on the database of the Italian Association of Blood Volunteers (AVIS), section of Ferrara, Italy, 2001–2010. Hematological data (170,238 exams referring to 16,422 donors) were categorized into seasonal and monthly intervals, and conventional and chronobiological analyses were applied. Results Platelets and plateletcrit were significantly higher in winter-autumn, with a main peak in December-February (average +3.4% and +4.6%, respectively, P <0.001 for both). Conclusions Although seasonal variations have been reported for several acute cardiovascular diseases, it is extremely unlikely that such a slight increase in platelet count in winter alone may be considered as a risk factor.
Collapse
Affiliation(s)
- Massimo Gallerani
- Internal Medicine, Azienda Ospedaliera-Universitaria, Ferrara, Italy.
| | | | | | | | | |
Collapse
|
85
|
Abstract
BACKGROUND Sepsis is a major disease affecting almost all organs and systems. OBJECTIVES To examine platelet count and indices (mean platelet volume (MPV) and platelet distribution width (PDW)) in severe sepsis. METHODS Patients with criteria for sepsis at a first examination by an Infectious Diseases specialist were selected. Consecutive patients who were admitted to the out-patient clinic and who were not diagnosed with any infectious disease were selected as the control group. RESULTS A total of 145 patients with sepsis and 143 patients as a control group were included in the study. MPV and PDW were significantly differentbetween sepsis patients and control group (P<0.05). Platelet count in sepsis patients was lower than control group but the difference was not significant. PDW was the unique significantly different parameter between survivors and non-survivors (p=0.001). CONCLUSION Platelet indices are important laboratory findings in the diagnosis of sepsis and severe sepsis. Severe sepsis patients who have greater than 18 % PDW levels have a higher risk of death. Therefore, PDW, which is part of an inexpensive, easily accessible and routinely performed test for almost all patients admitted to health facilities may be used for predicting mortality.
Collapse
Affiliation(s)
- E Guclu
- Department of Infectious Diseases and Clinical Microbiology, Ministry of Health Sakarya University Training and Research Hospital, Sakarya, Turkey
| | | | | |
Collapse
|
86
|
Hyaluronic acid-human blood hydrogels for stem cell transplantation. Biomaterials 2012; 33:8026-33. [PMID: 22898181 DOI: 10.1016/j.biomaterials.2012.07.058] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Accepted: 07/26/2012] [Indexed: 10/28/2022]
Abstract
Tissue engineering-based approaches have the potential to improve stem cell engraftment by increasing cell delivery to the myocardium. Our objective was to develop and characterize a naturally-derived, autologous, biodegradable hydrogel in order to improve acute stem cell retention in the myocardium. HA-blood hydrogels (HA-BL) were synthesized by mixing in a 1:1(v/v) ratio, lysed whole blood and hyaluronic acid (HA), whose carboxyl groups were functionalized with N-hydroxysuccinimide (NHS) to yield HA succinimidyl succinate (HA-NHS). We performed physical characterization and measured survival/proliferation of cardiosphere-derived cells (CDCs) encapsulated in the hydrogels. Hydrogels were injected intra-myocardially or applied epicardially in rats. NHS-activated carboxyl groups in HA react with primary amines present in blood and myocardium to form amide bonds, resulting in a 3D hydrogel bound to tissue. HA-blood hydrogels had a gelation time of 58±12 s, swelling ratio of 10±0.5, compressive and elastic modulus of 14±3 and 1.75±0.6 kPa respectively. These hydrogels were not degraded at 4 wks by hydrolysis alone. CDC encapsulation promoted their survival and proliferation. Intra-myocardial injection of CDCs encapsulated in these hydrogels greatly increased acute myocardial retention (p=0.001). Epicardial application of HA-blood hydrogels improved left ventricular ejection fraction following myocardial infarction (p=0.01). HA-blood hydrogels are highly adhesive, biodegradable, promote CDC survival and increase cardiac function following epicardial application after myocardial infarction.
Collapse
|
87
|
Lisman T, Pittau G, Leite FJT, De Boer MT, Meijer K, Kluin-Nelemans HC, Huls G, Te Boome LCJ, Kuball J, Nowak G, Fan ST, Azoulay D, Porte RJ. The circulating platelet count is not dictated by the liver, but may be determined in part by the bone marrow: analyses from human liver and stem cell transplantations. J Thromb Haemost 2012; 10:1624-30. [PMID: 22642442 DOI: 10.1111/j.1538-7836.2012.04800.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND The platelet count varies considerably between individuals, but within an individual the platelet count is remarkably stable over time. Mechanisms controlling the platelet count are not yet established. OBJECTIVE In the present study, we tested the hypothesis that the liver is important in controlling the circulating platelet count, as the liver is the main producer of thrombopoietin. METHODS We compared the platelet count prior to and after liver transplantation in >250 patients transplanted for familial amyloidotic polyneuropathy (FAP). In contrast to most patients undergoing liver transplantation, patients with FAP have normal liver function before transplantation. Furthermore, we compared platelet counts in 89 living liver donors with the platelet count in the recipients of these grafts. Finally we compared platelet counts in donor-recipient pairs of hematopoietic stem cells. RESULTS AND CONCLUSIONS The platelet count prior to transplantation correlated with the platelet count at 3 or 12 months after transplantation in patients with FAP (r=0.48, P<0.0001 at 3 months, r=0.39, P<0.0001 at 12 months), whereas the platelet count in a living liver donor did not correlate with the platelet count in the recipient at 3 or 12 months after transplantation (r=0.16, P=0.26 at 3 months, r=0.11, P=0.30 at 12 months). The platelet count of related donors of hematopoietic stem cells correlated with the platelet count in the recipient after transplantation (r=0.25, P=0.011). CONCLUSIONS These results suggest that the liver, in spite of being the prime producer of thrombopoietin, does not dictate the circulating platelet count, whereas the bone marrow does appear to play a role.
Collapse
Affiliation(s)
- T Lisman
- Section of Hepatobiliairy Surgery and Liver Transplantation, Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Paim CB, Paim FC, Da Silva AS, França RT, Costa MM, Leal CA, Soares JF, Labruna MB, Schetinger MR, Mazzanti A, Mazzanti CM, Monteiro SG, Lopes ST. Thrombocytopenia and platelet activity in dogs experimentally infected with Rangelia vitalii. Vet Parasitol 2012; 185:131-7. [DOI: 10.1016/j.vetpar.2011.09.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 09/26/2011] [Accepted: 09/28/2011] [Indexed: 11/29/2022]
|